Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Invest Ophthalmol Vis Sci ; 65(8): 9, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38958967

ABSTRACT

Purpose: Light detection destroys the visual pigment. Its regeneration, necessary for the recovery of light sensitivity, is accomplished through the visual cycle. Release of all-trans retinal by the light-activated visual pigment and its reduction to all-trans retinol comprise the first steps of the visual cycle. In this study, we determined the kinetics of all-trans retinol formation in human rod and cone photoreceptors. Methods: Single living rod and cone photoreceptors were isolated from the retinas of human cadaver eyes (ages 21 to 90 years). Formation of all-trans retinol was measured by imaging its outer segment fluorescence (excitation, 360 nm; emission, >420 nm). The extent of conversion of released all-trans retinal to all-trans retinol was determined by measuring the fluorescence excited by 340 and 380 nm. Measurements were repeated with photoreceptors isolated from Macaca fascicularis retinas. Experiments were carried out at 37°C. Results: We found that ∼80% to 90% of all-trans retinal released by the light-activated pigment is converted to all-trans retinol, with a rate constant of 0.24 to 0.55 min-1 in human rods and ∼1.8 min-1 in human cones. In M. fascicularis rods and cones, the rate constants were 0.38 ± 0.08 min-1 and 4.0 ± 1.1 min-1, respectively. These kinetics are several times faster than those measured in other vertebrates. Interphotoreceptor retinoid-binding protein facilitated the removal of all-trans retinol from human rods. Conclusions: The first steps of the visual cycle in human photoreceptors are several times faster than in other vertebrates and in line with the rapid recovery of light sensitivity exhibited by the human visual system.


Subject(s)
Macaca fascicularis , Retinal Cone Photoreceptor Cells , Retinal Rod Photoreceptor Cells , Vitamin A , Humans , Retinal Cone Photoreceptor Cells/physiology , Retinal Cone Photoreceptor Cells/metabolism , Aged , Retinal Rod Photoreceptor Cells/physiology , Aged, 80 and over , Middle Aged , Adult , Vitamin A/metabolism , Animals , Young Adult , Male , Retinaldehyde/metabolism , Cadaver , Female , Vision, Ocular/physiology , Retinal Pigments/metabolism
2.
Exp Eye Res ; 224: 109250, 2022 11.
Article in English | MEDLINE | ID: mdl-36122624

ABSTRACT

Sphingomyelinases (SMase), enzymes that catalyze the hydrolysis of sphingomyelin to ceramide, are important sensors for inflammatory cytokines and apoptotic signaling. Studies have provided evidence that increased SMase activity can contribute to retinal injury. In most tissues, two major SMases are responsible for stress-induced increases in ceramide: acid sphingomyelinase (ASMase) and Mg2+-dependent neutral sphingomyelinase (NSMase). The purposes of the current study were to determine the localization of SMases and their substrates in the retina and optic nerve head and to investigate the effects of ocular hypertension and ischemia on ASMase and NSMase activities. Tissue and cellular localization of ASMase and NSMase were determined by immunofluorescence imaging. Tissue localization of sphingomyelin in retinas was further determined by Matrix-Assisted Laser Desorption/Ionization mass spectrometry imaging. Tissue levels of sphingomyelins and ceramide were determined by liquid chromatography with tandem mass spectrometry. Sphingomyelinase activities under basal conditions and following acute ischemic and ocular hypotensive stress were measured using the Amplex Red Sphingomyelinase Assay Kit. Our data show that ASMase is in the optic nerve head and the retinal ganglion cell layer. NSMase is in the optic nerve head, photoreceptor and retinal ganglion cell layers. Both ASMase and NSMase were identified in human induced pluripotent stem cell-derived retinal ganglion cells and optic nerve head astrocytes. The retina and optic nerve head each exhibited unique distribution of sphingomyelins with the abundance of very long chain species being higher in the optic nerve head than in the retina. Basal activities for ASMase in retinas and optic nerve heads were 54.98 ± 2.5 and 95.6 ± 19.5 mU/mg protein, respectively. Ocular ischemia significantly increased ASMase activity to 86.2 ± 15.3 mU/mg protein in retinas (P = 0.03) but not in optic nerve heads (81.1 ± 15.3 mU/mg protein). Ocular hypertension significantly increased ASMase activity to 121.6 ± 7.3 mU/mg protein in retinas (P < 0.001) and 267.0 ± 66.3 mU/mg protein in optic nerve heads (P = 0.03). Basal activities for NSMase in retinas and optic nerve heads were 12.3 ± 2.1 and 37.9 ± 8.7 mU/mg protein, respectively. No significant change in NSMase activity was measured following ocular ischemia or hypertension. Our results provide evidence that both ASMase and NSMase are expressed in retinas and optic nerve heads; however, basal ASMase activity is significantly higher than NSMase activity in retinas and optic nerve heads. In addition, only ASMase activity was significantly increased in ocular ischemia or hypertension. These data support a role for ASMase-mediated sphingolipid metabolism in the development of retinal ischemic and hypertensive injuries.


Subject(s)
Hypertension , Induced Pluripotent Stem Cells , Ocular Hypertension , Optic Disk , Humans , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism , Sphingomyelins/pharmacology , Optic Disk/metabolism , Induced Pluripotent Stem Cells/metabolism , Retina/metabolism , Ceramides/metabolism , Cytokines , Ischemia
3.
Exp Eye Res ; 211: 108762, 2021 10.
Article in English | MEDLINE | ID: mdl-34499916

ABSTRACT

Ceramides are bioactive compounds that play important roles in regulating cellular responses to extracellular stimuli and stress. Previous studies have shown that ceramides contribute to retinal degeneration associated with ischemic and ocular hypertensive stress. Acid sphingomyelinase (ASMase) is one of the major enzymes responsible for the stress-induced generation of ceramides. The goals of this study are to investigate the effects of ceramides on retinal ganglion cells (RGCs) and of ASMase inhibition in ocular hypertensive mice. Induced pluripotent stem cell (iPSC)-derived RGCs and primary cultures of human optic nerve head astrocytes were used to characterize the response to C2-ceramide. Microbead-induced ocular hypertension in the ASMase heterozygote mouse model was used to confirm the physiological relevance of in vitro studies. In mice, RGC function and morphology were assessed with pattern ERG (pERG) and immunofluorescence. The addition of C2-ceramide to iPSC-derived RGCs produced a significant concentration- and time-dependent reduction in cell numbers when compared to control cultures. While the addition of C2-ceramide to astrocytes did not affect viability, it resulted in a 2.6-fold increase in TNF-α secretion. The addition of TNF-α or conditioned media from C2-ceramide-treated astrocytes to RGC cultures significantly reduced cell numbers by 56.1 ± 8.4% and 24.7 ± 4.8%, respectively. This cytotoxic response to astrocyte-conditioned media was blocked by TNF-α antibody. In ASMase heterozygote mice, functional and morphological analyses of ocular hypertensive eyes reveal significantly less RGC degeneration when compared with hypertensive eyes from wild-type mice. These results provide evidence that ceramides can induce RGC cell death by acting directly, as well as indirectly via the secretion of TNF-α from optic nerve head astrocytes. In vivo studies in mice provide evidence that ceramides derived through the activity of ASMase contribute to ocular hypertensive injury. Together these results support the importance of ceramides in the pathogenesis of ocular hypertensive injury to the retina.


Subject(s)
Ceramides/toxicity , Retinal Degeneration/chemically induced , Retinal Ganglion Cells/drug effects , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Cell Count , Cell Death , Electroretinography , Humans , Induced Pluripotent Stem Cells , Intraocular Pressure , Mice, Inbred C57BL , Mice, Knockout , Ocular Hypertension/metabolism , Optic Disk/cytology , Real-Time Polymerase Chain Reaction , Retinal Degeneration/metabolism , Retinal Ganglion Cells/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Invest Ophthalmol Vis Sci ; 62(2): 1, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33523199

ABSTRACT

Purpose: Bis-retinoids are a major component of lipofuscin that accumulates in the retinal pigment epithelium (RPE) in aging and age-related macular degeneration (AMD). Although bis-retinoids are known to originate from retinaldehydes required for the light response of photoreceptor cells, the relative contributions of the chromophore, 11-cis retinal, and photoisomerization product, all-trans retinal, are unknown. In photoreceptor outer segments, all-trans retinal, but not 11-cis retinal, is reduced by retinol dehydrogenase 8 (RDH8). Using Rdh8-/- mice, we evaluated the contribution of increased all-trans retinal to the formation and stability of RPE lipofuscin. Methods: Rdh8-/- mice were reared in cyclic-light or darkness for up to 6 months, with selected light-reared cohorts switched to dark-rearing for the final 1 to 8 weeks. The bis-retinoid A2E was measured from chloroform-methanol extracts of RPE-choroid using HPLC-UV/VIS spectroscopy. Lipofuscin fluorescence was measured from whole flattened eyecups (excitation, 488 nm; emission, 565-725 nm). Results: Cyclic-light-reared Rdh8-/- mice accumulated A2E and RPE lipofuscin approximately 1.5 times and approximately 2 times faster, respectively, than dark-reared mice. Moving Rdh8-/- mice from cyclic-light to darkness resulted in A2E levels less than expected to have accumulated before the move. Conclusions: Our findings establish that elevated levels of all-trans retinal present in cyclic-light-reared Rdh8-/- mice, which remain low in wild-type mice, contribute only modestly to RPE lipofuscin formation and accumulation. Furthermore, decreases in A2E levels occurring after moving cyclic-light-reared Rdh8-/- mice to darkness are consistent with processing of A2E within the RPE and the existence of a mechanism that could be a therapeutic target for controlling A2E cytotoxicity.


Subject(s)
Lipofuscin/metabolism , Macular Degeneration/metabolism , Retinal Pigment Epithelium/metabolism , Retinaldehyde/metabolism , Retinoids/metabolism , Animals , Chromatography, High Pressure Liquid , Disease Models, Animal , Female , Macular Degeneration/pathology , Male , Mice , Retinal Pigment Epithelium/pathology
5.
Photochem Photobiol Sci ; 19(10): 1300-1307, 2020 Oct 14.
Article in English | MEDLINE | ID: mdl-32812970

ABSTRACT

Retinal, the vitamin A aldehyde, is a potent photosensitizer that plays a major role in light-induced damage to vertebrate photoreceptors. 11-Cis retinal is the light-sensitive chromophore of rhodopsin, the photopigment of vertebrate rod photoreceptors. It is isomerized by light to all-trans, activating rhodopsin and beginning the process of light detection. All-trans retinal is released by activated rhodopsin, allowing its regeneration by fresh 11-cis retinal continually supplied to photoreceptors. The released all-trans retinal is reduced to all-trans retinol in a reaction using NADPH. We have examined the photooxidation mediated by 11-cis and all-trans retinal in single living rod photoreceptors isolated from mouse retinas. Photooxidation was measured with fluorescence imaging from the oxidation of internalized BODIPY C11, a fluorescent dye whose fluorescence changes upon oxidation. We found that photooxidation increased with the concentration of exogenously added 11-cis or all-trans retinal to metabolically compromised rod outer segments that lacked NADPH supply. In dark-adapted metabolically intact rod outer segments with access to NADPH, there was no significant increase in photooxidation following exposure of the cell to light, but there was significant increase following addition of exogenous 11-cis retinal. The results indicate that both 11-cis and all-trans retinal can mediate light-induced damage in rod photoreceptors. In metabolically intact cells, the removal of the all-trans retinal generated by light through its reduction to retinol minimizes all-trans retinal-mediated photooxidation. However, because the enzymatic machinery of the rod outer segment cannot remove 11-cis retinal, 11-cis-retinal-mediated photooxidation may play a significant role in light-induced damage to photoreceptor cells.


Subject(s)
Photoreceptor Cells/chemistry , Retinaldehyde/chemistry , Rod Cell Outer Segment/chemistry , Vitamin A/chemistry , Animals , Mice , Mice, Knockout , Molecular Structure , Optical Imaging , Oxidation-Reduction , Photochemical Processes
6.
Med Image Anal ; 56: 96-109, 2019 08.
Article in English | MEDLINE | ID: mdl-31203169

ABSTRACT

Autofluorescence is the emission of light by naturally occurring tissue components on the absorption of incident light. Autofluorescence within the eye is associated with several disorders, such as Age-related Macular Degeneration (AMD) which is a leading cause of central vision loss. Its pathogenesis is incompletely understood, but endogenous fluorophores in retinal tissue might play a role. Hyperspectral fluorescence microscopy of ex-vivo retinal tissue can be used to determine the fluorescence emission spectra of these fluorophores. Comparisons of spectra in healthy and diseased tissues can provide important insights into the pathogenesis of AMD. However, the spectrum from each pixel of the hyperspectral image is a superposition of spectra from multiple overlapping tissue components. As spectra cannot be negative, there is a need for a non-negative blind source separation model to isolate individual spectra. We propose a tensor formulation by leveraging multiple excitation wavelengths to excite the tissue sample. Arranging images from different excitation wavelengths as a tensor, a non-negative tensor decomposition can be performed to recover a provably unique low-rank model with factors representing emission and excitation spectra of these materials and corresponding abundance maps of autofluorescent substances in the tissue sample. We iteratively impute missing values common in fluorescence measurements using Expectation-Maximization and use L2 regularization to reduce ill-posedness. Further, we present a framework for performing group hypothesis testing on hyperspectral images, finding significant differences in spectra between AMD and control groups in the peripheral macula. In the absence of ground truth, i.e. molecular identification of fluorophores, we provide a rigorous validation of chosen methods on both synthetic and real images where fluorescence spectra are known. These methodologies can be applied to the study of other pathologies presenting autofluorescence that can be captured by hyperspectral imaging.


Subject(s)
Macular Degeneration/diagnostic imaging , Unsupervised Machine Learning , Humans , Imaging, Three-Dimensional , Macular Degeneration/pathology , Microscopy, Fluorescence , Models, Statistical
7.
Am J Physiol Gastrointest Liver Physiol ; 315(5): G713-G721, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30024770

ABSTRACT

Loss of retinyl ester (RE)-rich lipid droplets (LDs) from hepatic stellate cells (HSCs) is cited as a key event in their cellular transdifferentiation to activated, pro-fibrotic myofibroblasts; however, it remains unclear if changes in LD morphology or RE content are causal for transdifferentiation. To better understand LD dynamics in vitro within a common model of HSC activation, we used novel approaches preserving LD morphology and allowing for quantitation of RE. The size and quantity of LDs within in vitro and in vivo bile duct ligation (BDL)-activated HSCs were quantitated using adipocyte differentiation-related protein (ADRP) labeling and oil red o (ORO) staining (gold standard), and RE content was determined using fluorescence microscopy. We found during HSC activation in vitro that LD number differed significantly when measured by ADRP and ORO, respectively ( day 1: 56 vs. 5, P = 0.03; day 4: 101 vs. 39, P = 0.03; day 14: 241 vs. 12, P = 0.02). Ex vivo HSCs activated in vivo contained the same number of LDs as day 4 in vitro activated HSCs (118 vs. 101, P = 0.54). Decline in LD RE occurred beyond day 4 in vitro and day 1 ex vivo , after HSC transdifferentiation was underway. Lastly, in situ HSCs examined using electron microscopy show LDs tend to be smaller but are ultimately retained in BDL injured livers. Therefore, we conclude that during HSC transdifferentiation, LDs are not lost but are retained, decreasing in size. Additionally, RE content declines after transdifferentiation is underway. These data suggest that these LD changes are not causal for HSC transdifferentiation. NEW & NOTEWORTHY Loss of retinoid-laden lipid droplets from hepatic stellate cells has long been held as a hallmark of their transdifferentiation into activated myofibroblasts, the dominant cells that drive hepatic fibrosis. This study demonstrates that stellate cells activated in culture and after liver injury in vivo retain their lipid droplets and that these droplets become smaller and more numerous, with decreases in droplet retinoid concentration occurring only after cellular transdifferentiation is underway.


Subject(s)
Cell Transdifferentiation , Hepatic Stellate Cells/metabolism , Lipid Droplets/metabolism , Myofibroblasts/metabolism , Retinoids/metabolism , Animals , Cells, Cultured , Hepatic Stellate Cells/cytology , Male , Myofibroblasts/cytology , Rats , Rats, Sprague-Dawley
8.
Sci Rep ; 7(1): 17352, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29229934

ABSTRACT

Stargardt disease is a juvenile onset retinal degeneration, associated with elevated levels of lipofuscin and its bis-retinoid components, such as N-retinylidene-N-retinylethanolamine (A2E). However, the pathogenesis of Stargardt is still poorly understood and targeted treatments are not available. Utilizing high spatial and high mass resolution matrix assisted laser desorption ionization (MALDI) imaging mass spectrometry (IMS), we determined alterations of lipid profiles specifically localized to the retinal pigment epithelium (RPE) in Abca4 -/- Stargardt model mice compared to their relevant background strain. Extensive analysis by LC-MS/MS in both positive and negative ion mode was required to accurately confirm the identity of one highly expressed lipid class, bis(monoacylgylercoro)phosphate (BMP) lipids, and to distinguish them from isobaric species. The same BMP lipids were also detected in the RPE of healthy human retina. BMP lipids have been previously associated with the endosomal/lysosomal storage diseases Niemann-Pick and neuronal ceroid lipofuscinosis and have been reported to regulate cholesterol levels in endosomes. These results suggest that perturbations in lipid metabolism associated with late endosomal/lysosomal dysfunction may play a role in the pathogenesis of Stargardt disease and is evidenced in human retinas.


Subject(s)
Endosomes/pathology , Lipids/analysis , Lysophospholipids/metabolism , Lysosomes/pathology , Macular Degeneration/congenital , Monoglycerides/metabolism , Retina/pathology , Retinal Pigment Epithelium/pathology , ATP-Binding Cassette Transporters/physiology , Animals , Disease Models, Animal , Endosomes/metabolism , Humans , Lysosomes/metabolism , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mice , Mice, Knockout , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Stargardt Disease
9.
J Biol Chem ; 292(47): 19356-19365, 2017 11 24.
Article in English | MEDLINE | ID: mdl-28972139

ABSTRACT

Interphotoreceptor retinoid-binding protein (IRBP) is a specialized lipophilic carrier that binds the all-trans and 11-cis isomers of retinal and retinol, and this facilitates their transport between photoreceptors and cells in the retina. One of these retinoids, all-trans-retinal, is released in the rod outer segment by photoactivated rhodopsin after light excitation. Following its release, all-trans-retinal is reduced by the retinol dehydrogenase RDH8 to all-trans-retinol in an NADPH-dependent reaction. However, all-trans-retinal can also react with outer segment components, sometimes forming lipofuscin precursors, which after conversion to lipofuscin accumulate in the lysosomes of the retinal pigment epithelium and display cytotoxic effects. Here, we have imaged the fluorescence of all-trans-retinol, all-trans-retinal, and lipofuscin precursors in real time in single isolated mouse rod photoreceptors. We found that IRBP removes all-trans-retinol from individual rod photoreceptors in a concentration-dependent manner. The rate constant for retinol removal increased linearly with IRBP concentration with a slope of 0.012 min-1 µm-1 IRBP also removed all-trans-retinal, but with much less efficacy, indicating that the reduction of retinal to retinol promotes faster clearance of the photoisomerized rhodopsin chromophore. The presence of physiological IRBP concentrations in the extracellular medium resulted in lower levels of all-trans-retinal and retinol in rod outer segments following light exposure. It also prevented light-induced lipofuscin precursor formation, but it did not remove precursors that were already present. These findings reveal an important and previously unappreciated role of IRBP in protecting the photoreceptor cells against the cytotoxic effects of accumulated all-trans-retinal.


Subject(s)
Eye Proteins/physiology , Lipofuscin/metabolism , Retinaldehyde/metabolism , Retinol-Binding Proteins/physiology , Rod Cell Outer Segment/metabolism , Vitamin A/metabolism , Animals , Cattle , Light , Mice , Mice, Knockout
10.
Photochem Photobiol ; 93(3): 844-848, 2017 05.
Article in English | MEDLINE | ID: mdl-28500718

ABSTRACT

The RPE65 protein of the retinal pigment epithelium (RPE) enables the conversion of retinyl esters to the visual pigment chromophore 11-cis retinal. Fresh 11-cis retinal is generated from retinyl esters following photoisomerization of the visual pigment chromophore to all-trans during light detection. Large amounts of esters accumulate in Rpe65-/- mice, indicating their continuous formation when 11-cis retinal generation is blocked. We hypothesized that absence of light, by limiting the conversion of esters to 11-cis retinal, would also result in the build-up of retinyl esters in the RPE of wild-type mice. We used HPLC to quantify ester levels in organic extracts of the RPE from wild-type and Rpe65-/- mice. Retinyl ester levels in Sv/129 wild-type mice that were dark adapted for various intervals over a 4-week period were similar to those in mice raised in cyclic light. In C57BL/6 mice however, which contain less Rpe65 protein, dark adaptation was accompanied by an increase in ester levels compared to cyclic light controls. Retinyl ester levels were much higher in Rpe65-/- mice compared to wild type and kept increasing with age. The results suggest that the RPE65 role in retinyl ester homeostasis extends beyond enabling the formation of 11-cis retinal.


Subject(s)
Retinal Pigment Epithelium/metabolism , cis-trans-Isomerases/metabolism , Animals , Chromatography, High Pressure Liquid , Esters/metabolism , Mice, Inbred C57BL
11.
Exp Eye Res ; 155: 121-127, 2017 02.
Article in English | MEDLINE | ID: mdl-28219732

ABSTRACT

The accumulation of lipofuscin in the cells of the retinal pigment epithelium (RPE) is thought to play an important role in the development and progression of degenerative diseases of the retina. The bulk of RPE lipofuscin originates in reactions of the rhodopsin chromophore, retinal, with components of the photoreceptor outer segment. The 11-cis retinal isomer is generated in the RPE and supplied to rod photoreceptor outer segments where it is incorporated as the chromophore of rhodopsin. It is photoisomerized during light detection to all-trans and subsequently released by photoactivated rhodopsin as all-trans retinal, which is removed through reduction to all-trans retinol in a reaction requiring metabolic input in the form of NADPH. Both 11-cis and all-trans retinal can form lipofuscin precursor fluorophores in rod photoreceptor outer segments. Increased accumulation of lipofuscin has been suggested to result from excess formation of lipofuscin precursors due to buildup of all-trans retinal released by light exposure. In connection with this suggestion, the Abca4 transporter protein, an outer segment protein defects in which result in recessive Stargardt disease, has been proposed to promote the removal of all-trans retinal by facilitating its availability for reduction. To examine this possibility, we have measured the outer segment levels of all-trans retinal, all-trans retinol, and of lipofuscin precursors after bleaching by imaging the fluorescence of single rod photoreceptors isolated from wild type and Abca4-/- mice. We found that all-trans retinol and all-trans retinal levels increased after bleaching in both wild type and Abca4-/- rods. At all times after bleaching, there was no significant difference in all-trans retinal levels between the two strains. All-trans retinol levels were not significantly different between the two strains at early times, but were lower in Abca4-/- rods at times longer than 20 min after bleaching. Bleaching in the presence of lower metabolic substrate concentrations resulted in higher all-trans retinal levels and increased formation of lipofuscin precursors in both wild type and Abca4-/- rods. The results show that conditions that result in buildup of all-trans retinal levels result in increased generation of lipofuscin precursors in both wild type and Abca4-/- rods. The results are consistent with the proposal that Abca4 facilitates the reduction of all-trans retinal to retinol; absence of Abca4 however does not appear to be associated with higher all-trans retinal levels compared to wild type.


Subject(s)
Lipofuscin/metabolism , Retinal Degeneration/metabolism , Rod Cell Outer Segment/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Cells, Cultured , DNA/genetics , Disease Models, Animal , Mice , Mice, Transgenic , Mutation , Optical Imaging , Retinal Degeneration/pathology , Rod Cell Outer Segment/radiation effects , Ultraviolet Rays
12.
J Gen Physiol ; 148(1): 1-11, 2016 07.
Article in English | MEDLINE | ID: mdl-27353443

ABSTRACT

Photoactivation of vertebrate rhodopsin converts it to the physiologically active Meta II (R*) state, which triggers the rod light response. Meta II is rapidly inactivated by the phosphorylation of C-terminal serine and threonine residues by G-protein receptor kinase (Grk1) and subsequent binding of arrestin 1 (Arr1). Meta II exists in equilibrium with the more stable inactive form of rhodopsin, Meta III. Dark adaptation of rods requires the complete thermal decay of Meta II/Meta III into opsin and all-trans retinal and the subsequent regeneration of rhodopsin with 11-cis retinal chromophore. In this study, we examine the regulation of Meta III decay by Grk1 and Arr1 in intact mouse rods and their effect on rod dark adaptation. We measure the rates of Meta III decay in isolated retinas of wild-type (WT), Grk1-deficient (Grk1(-/-)), Arr1-deficient (Arr1(-/-)), and Arr1-overexpressing (Arr1(ox)) mice. We find that in WT mouse rods, Meta III peaks ∼6 min after rhodopsin activation and decays with a time constant (τ) of 17 min. Meta III decay slows in Arr1(-/-) rods (τ of ∼27 min), whereas it accelerates in Arr1(ox) rods (τ of ∼8 min) and Grk1(-/-) rods (τ of ∼13 min). In all cases, regeneration of rhodopsin with exogenous 11-cis retinal is rate limited by the decay of Meta III. Notably, the kinetics of rod dark adaptation in vivo is also modulated by the levels of Arr1 and Grk1. We conclude that, in addition to their well-established roles in Meta II inactivation, Grk1 and Arr1 can modulate the kinetics of Meta III decay and rod dark adaptation in vivo.


Subject(s)
Arrestins/metabolism , Dark Adaptation/physiology , G-Protein-Coupled Receptor Kinase 1/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Arrestins/genetics , G-Protein-Coupled Receptor Kinase 1/genetics , Mice , Mice, Knockout , Phosphorylation , Photic Stimulation , Protein Binding , Rhodopsin/metabolism
13.
Photochem Photobiol Sci ; 14(11): 1983-90, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26323192

ABSTRACT

The bis-retinoid N-retinylidene-N-retinylethanolamine (A2E) is one of the major components of lipofuscin, a fluorescent material that accumulates with age in the lysosomes of the retinal pigment epithelium (RPE) of the human eye. Lipofuscin, as well as A2E, exhibit a range of cytotoxic properties, which are thought to contribute to the pathogenesis of degenerative diseases of the retina such as Age-related Macular Degeneration. Consistent with such a pathogenic role, high levels of lipofuscin fluorescence are found in the central area of the human RPE, and decline toward the periphery. Recent reports have however suggested a surprising incongruence between the distributions of lipofuscin and A2E in the human RPE, with A2E levels being lowest in the central area and increasing toward the periphery. To appraise such a possibility, we have quantified the levels of A2E in the central and peripheral RPE areas of 10 eyes from 6 human donors (ages 75-91 years) with HPLC and UV/VIS spectroscopy. The levels of A2E in the central area were on average 3-6 times lower than in peripheral areas of the same eye. Furthermore, continuous accumulation of selected ions (CASI) imaging mass spectrometry showed the presence of A2E in the central RPE, and at lower intensities than in the periphery. We have therefore corroborated that in human RPE the levels of A2E are lower in the central area compared to the periphery. We conclude that the levels of A2E cannot by themselves provide an explanation for the higher lipofuscin fluorescence found in the central area of the human RPE.


Subject(s)
Retinal Pigment Epithelium/chemistry , Retinoids/analysis , Aged , Aged, 80 and over , Humans
14.
Prog Mol Biol Transl Sci ; 134: 449-63, 2015.
Article in English | MEDLINE | ID: mdl-26310170

ABSTRACT

Lipofuscin is highly fluorescent material, formed in several tissues but best studied in the eye. The accumulation of lipofuscin in the retinal pigment epithelium (RPE) is a hallmark of aging in the eye and has been implicated in various retinal degenerations, including age-related macular degeneration. The bis-retinoid N-retinyl-N-retinylidene ethanolamine (A2E), formed from retinal, has been identified as a byproduct of the visual cycle, and numerous in vitro studies have found toxicity associated with this compound. The compound is known to accumulate in the RPE with age and was the first identified compound extracted from lipofuscin. Our studies have correlated the distribution of lipofuscin and A2E across the human and mouse RPE. Lipofuscin fluorescence was imaged in the RPE from human donors of various ages and from assorted mouse models. The spatial distribution of A2E was determined using matrix-assisted laser desorption-ionization imaging mass spectrometry on both flat-mounted and transversally sectioned RPE tissue. Our data support the clinical observations in humans of strong RPE fluorescence, increasing with age, in the central area of the RPE. However, there was no correlation between the distribution of A2E and lipofuscin, as the levels of A2E were highest in the far periphery and decreased toward the central region. Interestingly, in all the mouse models, A2E distribution and lipofuscin fluorescence correlate well. These data demonstrate that the accumulation of A2E is not responsible for the increase in lipofuscin fluorescence observed in the central RPE with aging in humans.


Subject(s)
Lipofuscin/metabolism , Retinoids/metabolism , Animals , Humans , Imaging, Three-Dimensional , Models, Biological , Retinoids/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
15.
Prog Mol Biol Transl Sci ; 134: e1-12, 2015.
Article in English | MEDLINE | ID: mdl-26310175

ABSTRACT

Lipofuscin is a fluorescent mixture of partially digested proteins and lipids that accumulates with age in the lysosomal compartment of the retinal pigment epithelium (RPE) of the eye. Because it has been found to have significant cytotoxic potential, lipofuscin is thought to play a role in retinal degeneration diseases including age-related macular degeneration and Stargardt disease, a form of juvenile macular degeneration. The only known components of lipofuscin are bis-retinoids, the condensation products of two molecules of retinal. The bulk of lipofuscin is thought to originate in the rod photoreceptor outer segments as a by-product of reactions involving the retinal chromophore of rhodopsin. 11-cis retinal flows from the RPE into the rod outer segments, where it combines with opsin to form rhodopsin; all-trans retinal is released into the rod outer segments by photoactivated rhodopsin following its excitation by light. Both 11-cis and all-trans retinal can generate lipofuscin-like fluorophores and bis-retinoids when added to rod outer segment membranes. The levels of lipofuscin precursor fluorophores present in the outer segments of dark-adapted rods are similar in cyclic-light- and dark-reared mice, as are the levels of accumulated lipofuscin in the RPE. Because the retinol dehydrogenase enzyme present in rod outer segments can reduce all-trans but not 11-cis retinal, lipofuscin precursors are more likely to form from 11-cis than all-trans retinal, even under cyclic light conditions. Thus, 11-cis retinal may be the primary source of lipofuscin in the retina.


Subject(s)
Lipofuscin/metabolism , Retina/metabolism , Retinaldehyde/metabolism , Animals , Humans , Models, Biological , Retinal Pigment Epithelium/metabolism , Rhodopsin/metabolism
16.
Photochem Photobiol Sci ; 14(10): 1888-95, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26223373

ABSTRACT

The accumulation of lipofuscin, an autofluorescent aging marker, in the retinal pigment epithelium (RPE) has been implicated in the development of age-related macular degeneration (AMD). Lipofuscin contains several visual cycle byproducts, most notably the bisretinoid N-retinylidene-N-retinylethanolamine (A2E). Previous studies with human donor eyes have shown a significant mismatch between lipofuscin autofluorescence (AF) and A2E distributions. The goal of the current project was to examine this relationship in a primate model with a retinal anatomy similar to that of humans. Ophthalmologically naive young (<10 years., N = 3) and old (>10 years., N = 4) Macaca fascicularis (macaque) eyes, were enucleated, dissected to yield RPE/choroid tissue, and flat-mounted on indium-tin-oxide-coated conductive slides. To compare the spatial distributions of lipofuscin and A2E, fluorescence and mass spectrometric imaging were carried out sequentially on the same samples. The distribution of lipofuscin fluorescence in the primate RPE reflected previously obtained human results, having the highest intensities in a perifoveal ring. Contrarily, A2E levels were consistently highest in the periphery, confirming a lack of correlation between the distributions of lipofuscin and A2E previously described in human donor eyes. We conclude that the mismatch between lipofuscin AF and A2E distributions is related to anatomical features specific to primates, such as the macula, and that this primate model has the potential to fill an important gap in current AMD research.


Subject(s)
Lipofuscin/metabolism , Retinal Pigment Epithelium/metabolism , Retinoids/metabolism , Aging/metabolism , Animals , Humans , Macaca
17.
PLoS One ; 10(7): e0133032, 2015.
Article in English | MEDLINE | ID: mdl-26168297

ABSTRACT

BACKGROUND: Mutations of acid sphingomyelinase (ASMase) cause Niemann-Pick diseases type A and B, which are fatal inherited lipid lysosomal storage diseases, characterized with visceral organ abnormalities and neurodegeneration. However, the effects of suppressing retinal ASMase expression are not understood. The goal of this study was to determine if the disruption of ASMase expression impacts the retinal structure and function in the mouse, and begin to investigate the mechanisms underlying these abnormalities. METHODS: Acid sphingomyelinase knockout (ASMase KO) mice were utilized to study the roles of this sphingolipid metabolizing enzyme in the retina. Electroretinogram and morphometric analysis were used to assess the retinal function and structure at various ages. Sphingolipid profile was determined by liquid chromatography-mass spectrometry. Western blots evaluated the level of the autophagy marker LC3-II. RESULTS: When compared to control animals, ASMase KO mice exhibited significant age-dependent reduction in ERG a- and b-wave amplitudes. Associated with these functional deficits, morphometric analysis revealed progressive thinning of retinal layers; however, the most prominent degeneration was observed in the photoreceptor and outer nuclear layer. Additional analyses of ASMase KO mice revealed early reduction in ERG c-wave amplitudes and increased lipofuscin accumulation in the retinal pigment epithelium (RPE). Sphingolipid analyses showed abnormal accumulation of sphingomyelin and sphingosine in ASMase KO retinas. Western blot analyses showed a higher level of the autophagosome marker LC3-II. CONCLUSIONS: These studies demonstrate that ASMase is necessary for the maintenance of normal retinal structure and function. The early outer retinal dysfunction, outer segment degeneration, accumulation of lipofuscin and autophagosome markers provide evidence that disruption of lysosomal function contributes to the age-dependent retinal degeneration exhibited by ASMase KO mice.


Subject(s)
Aging/pathology , Retinal Pigment Epithelium/pathology , Sphingomyelin Phosphodiesterase/metabolism , Aging/metabolism , Animals , Electroretinography , Mice , Mice, Inbred C57BL , Mice, Knockout , Retinal Pigment Epithelium/enzymology , Retinal Pigment Epithelium/physiopathology , Sphingomyelin Phosphodiesterase/genetics
18.
Methods Mol Biol ; 1271: 327-43, 2015.
Article in English | MEDLINE | ID: mdl-25697533

ABSTRACT

Absorption of light isomerizes the retinyl chromophore of the photoreceptor pigment rhodopsin from 11-cis to all-trans, generating the photoactivated rhodopsin form. The photoisomerization of the chromophore however destroys rhodopsin, and its regeneration requires the removal of the all-trans and the supply of fresh 11-cis chromophore. The all-trans chromophore is removed through a series of steps beginning with its release from photoactivated rhodopsin in the form of all-trans-retinal, leaving behind the apoprotein opsin. All-trans-retinal is then reduced to all-trans-retinol, which is transported out of the photoreceptor. Rhodopsin is regenerated from opsin and fresh 11-cis-retinal arriving to the photoreceptor from the retinal pigment epithelium. Both all-trans and 11-cis-retinal can form precursors of lipofuscin, a pigment that accumulates with age in the lysosomal compartment of the retinal pigment epithelium. All-trans-retinal, all-trans-retinol, and lipofuscin precursors all emit significant and distinct fluorescence signals, allowing their monitoring in single photoreceptor cells with fluorescence imaging. Here we describe the procedures for measuring these fluorophores in single mouse rod photoreceptors.


Subject(s)
Photoreceptor Cells/metabolism , Rhodopsin/metabolism , Animals , Lipofuscin/metabolism , Mice , Tretinoin/metabolism , Vitamin A/metabolism
19.
J Interprof Care ; 29(3): 253-5, 2015 May.
Article in English | MEDLINE | ID: mdl-25180629

ABSTRACT

For students interested in enriching their interprofessional competencies beyond those required and offered by their academic programs, an elective interprofessional education fellowship can serve that need. We designed a fellowship for students linking a conceptual framework grounded in adult learning principles. During the fellowship, students progress through three levels of learning as they acquire, apply, and demonstrate interprofessional collaborative knowledge and skills; fellowship activities are self-directed. A content analysis of students' fellowship summary reports sought to determine the effectiveness of the fellowship as a learning experience to acquire interprofessional collaborative competences. Results indicated that students most consistently report competencies associated with acquisition of values and ethics for interprofessional practice, roles/responsibilities, and teams/teamwork; interprofessional communication was implied. All students expressed commitment to interprofessional collaborative behavior when in practice. Based on the results from this study, this fellowship structure may serve as a model for other institutions to adapt and implement for best practice and best fit.


Subject(s)
Cooperative Behavior , Fellowships and Scholarships/organization & administration , Interprofessional Relations , Patient Care Team/organization & administration , Communication , Fellowships and Scholarships/standards , Humans , Patient Care Team/standards , Problem-Based Learning , Professional Competence , Professional Role
20.
J Am Soc Mass Spectrom ; 25(8): 1394-403, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24819461

ABSTRACT

Matrix assisted laser desorption ionization imaging mass spectrometry (MALDI IMS) has the ability to provide an enormous amount of information on the abundances and spatial distributions of molecules within biological tissues. The rapid progress in the development of this technology significantly improves our ability to analyze smaller and smaller areas and features within tissues. The mammalian eye has evolved over millions of years to become an essential asset for survival, providing important sensory input of an organism's surroundings. The highly complex sensory retina of the eye is comprised of numerous cell types organized into specific layers with varying dimensions, the thinnest of which is the 10 µm retinal pigment epithelium (RPE). This single cell layer and the photoreceptor layer contain the complex biochemical machinery required to convert photons of light into electrical signals that are transported to the brain by axons of retinal ganglion cells. Diseases of the retina, including age-related macular degeneration (AMD), retinitis pigmentosa, and diabetic retinopathy, occur when the functions of these cells are interrupted by molecular processes that are not fully understood. In this report, we demonstrate the use of high spatial resolution MALDI IMS and FT-ICR tandem mass spectrometry in the Abca4(-/-) knockout mouse model of Stargardt disease, a juvenile onset form of macular degeneration. The spatial distributions and identity of lipid and retinoid metabolites are shown to be unique to specific retinal cell layers.


Subject(s)
Lipids/analysis , Models, Biological , Retina/pathology , Retinoids/analysis , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Cyclotrons , Diagnostic Imaging , Fourier Analysis , Lipids/chemistry , Macular Degeneration/diagnosis , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mice, Inbred C57BL , Mice, Knockout , Photoreceptor Cells, Vertebrate/chemistry , Photoreceptor Cells, Vertebrate/pathology , Retina/chemistry , Retinal Neurons/chemistry , Retinal Neurons/pathology , Retinal Pigment Epithelium/chemistry , Retinal Pigment Epithelium/pathology , Retinoids/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stargardt Disease , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...