Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 18(4)2017 Mar 29.
Article in English | MEDLINE | ID: mdl-28353645

ABSTRACT

The accumulation of lipofuscin in the retinal pigment epithelium (RPE) is dependent on the effectiveness of photoreceptor outer segment material degradation. This study explored the role of autophagy in the fate of RPE lipofuscin degradation. After seven days of feeding with either native or modified rod outer segments, ARPE-19 cells were treated with enhancers or inhibitors of autophagy and the autofluorescence was detected by fluorescence-activated cell sorting. Supplementation with different types of rod outer segments increased lipofuscin-like autofluorescence (LLAF) after the inhibition of autophagy, while the induction of autophagy (e.g., application of rapamycin) decreased LLAF. The effects of autophagy induction were further confirmed by Western blotting, which showed the conversion of LC3-I to LC3-II, and by immunofluorescence microscopy, which detected the lysosomal activity of the autophagy inducers. We also monitored LLAF after the application of several autophagy inhibitors by RNA-interference and confocal microscopy. The results showed that, in general, the inhibition of the autophagy-related proteins resulted in an increase in LLAF when cells were fed with rod outer segments, which further confirms the effect of autophagy in the fate of RPE lipofuscin degradation. These results emphasize the complex role of autophagy in modulating RPE autofluorescence and confirm the possibility of the pharmacological clearance of RPE lipofuscin by small molecules.


Subject(s)
Autophagy , Lipofuscin/metabolism , Retinal Pigment Epithelium/metabolism , Cell Line , Fluorescence , Humans , Lysosomes/metabolism , Retinal Pigment Epithelium/drug effects , Sirolimus/pharmacology
2.
PLoS One ; 11(2): e0148773, 2016.
Article in English | MEDLINE | ID: mdl-26867008

ABSTRACT

PURPOSE: Bardet-Biedl syndrome is a complex ciliopathy that usually manifests with some form of retinal degeneration, amongst other ciliary-related deficiencies. One of the genetic causes of this syndrome results from a defect in Bardet-Biedl Syndrome 5 (BBS5) protein. BBS5 is one component of the BBSome, a complex of proteins that regulates the protein composition in cilia. In this study, we identify a smaller molecular mass form of BBS5 as a variant formed by alternative splicing and show that expression of this splice variant is restricted to the retina. METHODS: Reverse transcription PCR from RNA was used to isolate and identify potential alternative transcripts of Bbs5. A peptide unique to the C-terminus of the BBS5 splice variant was synthesized and used to prepare antibodies that selectively recognized the BBS5 splice variant. These antibodies were used on immunoblots of tissue extracts to determine the extent of expression of the alternative transcript and on tissue slices to determine the localization of expressed protein. Pull-down of fluorescently labeled arrestin1 by immunoprecipitation of the BBS5 splice variant was performed to assess functional interaction between the two proteins. RESULTS: PCR from mouse retinal cDNA using Bbs5-specific primers amplified a unique cDNA that was shown to be a splice variant of BBS5 resulting from the use of cryptic splicing sites in Intron 7. The resulting transcript codes for a truncated form of the BBS5 protein with a unique 24 amino acid C-terminus, and predicted 26.5 kD molecular mass. PCR screening of RNA isolated from various ciliated tissues and immunoblots of protein extracts from these same tissues showed that this splice variant was expressed in retina, but not brain, heart, kidney, or testes. Quantitative PCR showed that the splice variant transcript is 8.9-fold (+/- 1.1-fold) less abundant than the full-length transcript. In the retina, the splice variant of BBS5 appears to be most abundant in the connecting cilium of photoreceptors, where BBS5 is also localized. Like BBS5, the binding of BBS5L to arrestin1 can be modulated by phosphorylation through protein kinase C. CONCLUSIONS: In this study we have identified a novel splice variant of BBS5 that appears to be expressed only in the retina. The BBS5 splice variant is expressed at approximately 10% of full-length BBS5 level. No unique functional or localization properties could be identified for the splice variant compared to BBS5.


Subject(s)
Alternative Splicing , Bardet-Biedl Syndrome/genetics , Carrier Proteins/metabolism , Gene Expression Regulation , Proteins/metabolism , Retina/metabolism , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/genetics , Cattle , Cilia/metabolism , Cytoskeletal Proteins , DNA, Complementary/metabolism , Humans , Immunoprecipitation , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neurons/metabolism , Phosphate-Binding Proteins , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Proteins/genetics , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Sus scrofa , Tissue Distribution , Xenopus laevis
3.
Cell Mol Life Sci ; 70(23): 4603-16, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23817741

ABSTRACT

Arrestins are dynamic proteins that move between cell compartments triggered by stimulation of G-protein-coupled receptors. Even more dynamically in vertebrate photoreceptors, arrestin1 (Arr1) moves between the inner and outer segments according to the light conditions. Previous studies have shown that the light-driven translocation of Arr1 in rod photoreceptors is initiated by rhodopsin through a phospholipase C/protein kinase C (PKC) signaling cascade. The purpose of this study is to identify the PKC substrate that regulates the translocation of Arr1. Mass spectrometry was used to identify the primary phosphorylated proteins in extracts prepared from PKC-stimulated mouse eye cups, confirming the finding with in vitro phosphorylation assays. Our results show that Bardet-Biedl syndrome 5 (BBS5) is the principal protein phosphorylated either by phorbol ester stimulation or by light stimulation of PKC. Via immunoprecipitation of BBS5 in rod outer segments, Arr1 was pulled down; phosphorylation of BBS5 reduced this co-precipitation of Arr1. Immunofluorescence and immunoelectron microscopy showed that BBS5 principally localizes along the axonemes of rods and cones, but also in photoreceptor inner segments, and synaptic regions. Our principal findings in this study are threefold. First, we demonstrate that BBS5 is post-translationally regulated by phosphorylation via PKC, an event that is triggered by light in photoreceptor cells. Second, we find a direct interaction between BBS5 and Arr1, an interaction that is modulated by phosphorylation of BBS5. Finally, we show that BBS5 is distributed along the photoreceptor axoneme, co-localizing with Arr1 in the dark. These findings suggest a role for BBS5 in regulating light-dependent translocation of Arr1 and a model describing its role in Arr1 translocation is proposed.


Subject(s)
Arrestins/metabolism , Carrier Proteins/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Animals, Genetically Modified , Arrestins/genetics , Axoneme/metabolism , Carrier Proteins/genetics , Cytoskeletal Proteins , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoblotting , Light , Mass Spectrometry , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Immunoelectron , Models, Biological , Phosphate-Binding Proteins , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/radiation effects , Protein Binding/radiation effects , Protein Kinase C/metabolism , Retinal Rod Photoreceptor Cells/ultrastructure , Xenopus , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
4.
Exp Eye Res ; 112: 57-67, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23603319

ABSTRACT

The mechanisms that control the natural rate of lipofuscin accumulation in the retinal pigment epithelial (RPE) cell and its stability over time are not well understood. Similarly, the contributions of retinoids, phospholipids and oxidation to the rate of accumulation of lipofuscin are uncertain. The experiments in this study were conducted to explore the individual contribution of rod outer segments (ROS) components to lipofuscin formation and its accumulation and stability over time. During the period of 14 days incubation of ROS, lipofuscin-like autofluorescence (LLAF) determined at two wavelengths (530 and 585 nm) by fluorescence-activated cell sorting (FACS) was measured from RPE cells. The autofluorescence increased in an exponential manner with a strong linear component between days 1 and 7. The magnitude of the increase was larger in cells incubated with 4-hydroxynonenal (HNE-ROS) compared with cells incubated with either bleached or unbleached ROS, but with a different spectral profile. A small (10-15%) decrease in LLAF was observed after stopping the ROS feeding for 14 days. The phagocytosis rate of HNE-ROS was higher than that of either bleached or unbleached ROS during the first 24 h of supplementation. Among the different ROS components, the increase of LLAF was highest in cells incubated with all-trans-retinal. Surprisingly, incubation with 11-cis-retinal and 9-cis-retinal also resulted in strong LLAF increase, comparable to the increase induced by all-trans-retinal. Supplementation with liposomes containing phosphatidylethanolamine (22: 6-PE) and phosphatidylcholine (18:1-PC) also increased LLAF, while incubation with opsin had little effect. Cells incubated with retinoids demonstrated strong dose-dependence in LLAF increase, and the magnitude of the increase was 2-3 times higher at 585 nm compared to 530 nm, while cells incubated with liposomes showed little dose-dependence and similar increase at both wavelengths. Very little difference in LLAF was noted between cells incubated with either unbleached or bleached ROS under any conditions. In summary, results from this study suggest that supplementation with various ROS components can lead to an increase in LLAF, although the autofluorescence generated by the different classes of components has distinct spectral profiles, where the autofluorescence induced by retinoids results in a spectral profile closest to the one observed from human lipofuscin. Future fluorescence characterization of LLAF in vitro would benefit from an analysis of multiple wavelengths to better match the spectral characteristics of lipofuscin in vivo.


Subject(s)
Lipofuscin/metabolism , Retinal Pigment Epithelium/metabolism , Retinoids/pharmacology , Rod Cell Outer Segment/drug effects , Aldehydes/pharmacology , Animals , Cattle , Cell Line , Cells, Cultured , Diterpenes , Flow Cytometry , Humans , Liposomes , Microscopy, Confocal , Phagocytosis/physiology , Phosphatidylcholines/pharmacology , Phosphatidylethanolamines/pharmacology , Retinaldehyde/pharmacology , Rod Cell Outer Segment/metabolism , Rod Cell Outer Segment/radiation effects , Tretinoin/pharmacology
5.
J Immunol ; 187(4): 1977-85, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21765017

ABSTRACT

Noninfectious uveitis is a leading cause of blindness and thought to involve autoimmune T cell responses to retinal proteins (e.g., retinal arrestin [soluble-Ag (S-Ag)]). There are no known biomarkers for the disease. Susceptibility is associated with HLA, but little is known about susceptible class II alleles or the potentially pathogenic epitopes that they present. Using a humanized HLA-transgenic mouse model of S-Ag-induced autoimmune uveitis, we identified several susceptible and resistant alleles of HLA-DR and -DQ genes and defined pathogenic epitopes of S-Ag presented by the susceptible alleles. The sequences of these epitopes overlap with some previously identified peptides of S-Ag ("M" and "N"), known to elicit memory responses in lymphocytes of uveitis patients. HLA-DR-restricted, S-Ag-specific CD4(+) T cells could be detected in blood and draining lymph nodes of uveitic mice with HLA class II tetramers and transferred the disease to healthy mice. Importantly, tetramer-positive cells were detected in peripheral blood of a uveitis patient. To our knowledge, these findings provide the first tangible evidence that an autoimmune response to retina is causally involved in pathogenesis of human uveitis, demonstrating the feasibility of identifying and isolating retinal Ag-specific T cells from uveitis patients and may facilitate their development as biomarkers for the disease.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Eye Proteins/immunology , HLA-DQ Antigens/immunology , HLA-DR Antigens/immunology , Retina/immunology , Uveitis/immunology , Alleles , Animals , Autoantigens/genetics , Autoimmune Diseases/genetics , Biomarkers , CD4-Positive T-Lymphocytes/pathology , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Eye Proteins/genetics , Genetic Predisposition to Disease , HLA-DQ Antigens/genetics , HLA-DR Antigens/genetics , Humans , Immunologic Memory/genetics , Immunologic Memory/immunology , Mice , Mice, Transgenic , Retina/pathology , Uveitis/genetics , Uveitis/pathology
6.
Invest Ophthalmol Vis Sci ; 52(3): 1832-40, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21051714

ABSTRACT

PURPOSE: Arrestin is in disequilibrium in photoreceptors, translocating between inner and outer segments in response to light. The purpose of this project was to identify the cellular component with which arrestin associates in the dark-adapted retina. METHODS: Retinas were cross-linked with 2.5 mM dithiobis(succinimidylpropionate) (DSP), and arrestin-containing complexes purified by anion-exchange chromatography. Tandem mass spectrometric analysis was used to identify the protein components in the complex. Enolase localization in photoreceptors was assessed by immunohistochemistry. Confirmation of interacting components was performed using immunoprecipitation and surface plasmon resonance (SPR). Enolase activity was also assessed in the presence of arrestin1. RESULTS: In retinas treated with DSP, arrestin cross-linked in a 125-kDa complex. The principal components of this complex were arrestin1 and enolase1. Both arrestin1 and -4 were pulled down with enolase1 when enolase1 was immunoprecipitated. In the dark-adapted retina, enolase1 co-localized with arrestin1 in the inner segments and outer nuclear layer, but remained in the inner segments when arrestin1 translocated in response to light adaptation. SPR of purified arrestin1 and enolase1 demonstrated direct binding between arrestin1 and enolase1. Arrestin1 modulated the catalytic activity of enolase1, slowing it by as much as 24%. CONCLUSIONS: The results show that in the dark-adapted retina, arrestin1 and -4 interact with enolase1. The SPR data show that the interaction between arrestin1 and enolase1 was direct, not requiring a third element to form the complex. Arrestin1 slowed the catalytic activity of enolase1, suggesting that light-driven translocation of arrestin1 may modulate the metabolic activity of photoreceptors.


Subject(s)
Arrestin/metabolism , Phosphopyruvate Hydratase/metabolism , Photoreceptor Cells, Vertebrate/enzymology , Animals , Arrestin/pharmacology , Cattle , Chromatography, Ion Exchange , Cross-Linking Reagents , Dark Adaptation , Immunohistochemistry , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/radiation effects , Protein Binding , Surface Plasmon Resonance , Tandem Mass Spectrometry , Xenopus laevis
7.
Invest Ophthalmol Vis Sci ; 49(7): 3224-30, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18378578

ABSTRACT

PURPOSE: Many mutations in rhodopsin, including P23H, result in misfolding and mislocalization of the protein. It has been demonstrated that pharmacologic chaperones are effective in assisting the proper folding and targeting of P23H opsin. This study was designed to investigate a high-throughput screening strategy for identification of pharmacologic chaperones by using a combination of in silico, cell-based, and in vitro METHODS: methods. A library of 24,000 drug-like small molecules was screened by in silico molecular docking with DOCK5.1. The top hits were assayed in an in vitro competition assay. The selected compound was then assayed for pharmacologic chaperoning activity in stable cell lines expressing wild-type and P23H opsin. RESULTS: Beta-ionone was easily identified by the high-throughput screen. It strongly inhibits rhodopsin formation and, when incubated in cells expressing P23H opsin, resulted in a 2.5-fold rescue of P23H opsin. The screen also identified compound NSC45012 [1-(3,5-dimethyl-1H-pyrazol-4-yl)ethanone], a weak inhibitor of opsin regeneration and resulted in a 40% rescue of the mutant opsin. The level of rescue correlated well with the extent of inhibition. CONCLUSIONS: A combination of in silico and cell-based screening provides a useful tool for identifying pharmacologic chaperones for P23H opsin. This approach identified both potent and weak pharmacologic chaperones. Both types of molecules may be potential candidates for treatment of opsin-related RP.


Subject(s)
Molecular Chaperones/chemistry , Molecular Chaperones/pharmacology , Mutation , Protein Folding , Rhodopsin/chemistry , Rhodopsin/genetics , Animals , Cattle , Cell Line , Histidine , Molecular Weight , Norisoprenoids/pharmacology , Proline , Pyrazoles/pharmacology , Rhodopsin/drug effects
8.
Mol Vis ; 13: 1701-10, 2007 Sep 18.
Article in English | MEDLINE | ID: mdl-17960108

ABSTRACT

PURPOSE: Dramatic restoration of retinal function has followed subretinal viral-mediated gene therapy in RPE65-deficient animal models of human Leber congenital amaurosis (LCA) caused by RPE65 mutations. Progress in early-phase clinical trials of RPE65-LCA prompted us to begin development of an in vivo bioassay of clinical grade vector stability for later-phase trials. METHODS: Naturally-occurring Rpe65-mutant rd12 mice (2-4 mo of age) were studied with full-field electroretinograms (ERGs). Flash stimuli (range, -4.1 to 3.6 log scot-cd x s x m(-2)) were used to evoke ERGs in anesthetized, dark-adapted mice. B-wave amplitudes were measured conventionally and luminance-response functions were fit. Leading edges of photoresponses were analyzed with a model of rod phototransduction activation. A unilateral subretinal injection of AAV2-CB(SB)-hRPE65 vector was delivered and therapeutic efficacy of 4 doses spanning a 2 log unit range was studied with ERGs performed about 6 weeks after injection. Uninjected rd12 eyes and wild-type (wt) mice served as controls. RESULTS: Rd12 mice showed substantially smaller amplitudes and lower sensitivities than wt mice for all measured ERG b-wave and photoresponse parameters. For the dose-response study, there was no difference between 0.01X-dosed mice and untreated mutants. Improved receptoral and post-receptoral function was evident for 0.1X, 0.3X, 1X doses: b-wave semi-saturation constants decreased, b-wave amplitudes increased with dose; photoresponses showed faster kinetics and higher maximum amplitudes. ERG b-wave amplitude to a selected stimulus light intensity could provide evidence of biologic activity of the vector; interocular differences in b-wave amplitude comparing treated versus untreated eyes in the same animal also revealed vector efficacy. CONCLUSIONS: We have taken the first steps toward developing an ERG assay of biologic activity of human grade vector for future clinical trials of RPE65-LCA. Faithful murine models of treatable human disease tested with specific ERG protocols may emerge as valuable in vivo bioassays for future human clinical trials of therapy in many retinal degenerative diseases.


Subject(s)
Carrier Proteins/genetics , Electroretinography , Eye Proteins/genetics , Genetic Therapy , Mutation , Retinal Degeneration/genetics , Animals , Biological Assay , Blindness/congenital , Blindness/therapy , Clinical Trials as Topic , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Mutant Strains , Optic Atrophy, Hereditary, Leber/therapy , Photic Stimulation , Retinal Degeneration/diagnosis , Retinal Degeneration/physiopathology , Retinal Degeneration/therapy , cis-trans-Isomerases
9.
J Biol Chem ; 282(35): 25560-8, 2007 Aug 31.
Article in English | MEDLINE | ID: mdl-17606620

ABSTRACT

In this study we investigate conformational changes in Loop V-VI of visual arrestin during binding to light-activated, phosphorylated rhodopsin (Rho*-P) using a combination of site-specific cysteine mutagenesis and intramolecular fluorescence quenching. Introduction of cysteines at positions in the N-domain at residues predicted to be in close proximity to Ile-72 in Loop V-VI of arrestin (i.e. Glu-148 and Lys-298) appear to form an intramolecular disulfide bond with I72C, significantly diminishing the binding of arrestin to Rho*-P. Using a fluorescence approach, we show that the steady-state emission from a monobromobimane fluorophore in Loop V-VI is quenched by tryptophan residues placed at 148 or 298. This quenching is relieved upon binding of arrestin to Rho*-P. These results suggest that arrestin Loop V-VI moves during binding to Rho*-P and that conformational flexibility of this loop is essential for arrestin to adopt a high affinity binding state.


Subject(s)
Arrestin/chemistry , Rhodopsin/chemistry , Amino Acid Substitution , Animals , Arrestin/genetics , Arrestin/metabolism , Binding Sites , Bridged Bicyclo Compounds/chemistry , Bridged Bicyclo Compounds/metabolism , Cattle , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Humans , Models, Molecular , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Protein Structure, Secondary , Rhodopsin/genetics , Rhodopsin/metabolism , Spectrometry, Fluorescence
10.
Mol Ther ; 13(3): 565-72, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16223604

ABSTRACT

Retinal pigment epithelium-specific protein 65 kDa (RPE65) is a protein responsible for isomerization of all-trans-retinaldehyde to its photoactive 11-cis-retinaldehyde and is essential for the visual cycle. RPE65 mutations can cause severe, early onset retinal diseases such as Leber congenital amaurosis (LCA). A naturally occurring rodent model of LCA with a recessive nonsense Rpe65 mutation, the rd12 mouse, displays a profoundly diminished rod electroretinogram (ERG), an absence of 11-cis-retinaldehyde and rhodopsin, an overaccumulation of retinyl esters in retinal pigmented epithelial (RPE) cells, and photoreceptor degeneration. rd12 mice were injected subretinally at postnatal day 14 with rAAV5-CBA-hRPE65 vector. RPE65 expression was found over large areas of RPE soon after treatment. This led to improved rhodopsin levels with ERG signals restored to near normal. Retinyl ester levels were maintained at near normal, and fundus and retinal morphology remained normal. All parameters of restored retinal health remained stable for at least 7 months. The Morris water maze behavioral test was modified to test rod function under very dim light; rd12 mice treated in one eye performed similar to normally sighted C57BL/6J mice, while untreated rd12 mice performed very poorly, demonstrating that gene therapy can restore normal vision-dependent behavior in a congenitally blind animal.


Subject(s)
Carrier Proteins/genetics , Eye Proteins/genetics , Genetic Therapy , Optic Atrophy, Hereditary, Leber/genetics , Optic Atrophy, Hereditary, Leber/therapy , Retina/anatomy & histology , Retina/physiology , Vision, Ocular/genetics , Animals , Behavior, Animal/physiology , Dependovirus , Disease Models, Animal , Esters , Genetic Vectors , Mice , Mice, Inbred C57BL , Optic Atrophy, Hereditary, Leber/pathology , Retina/pathology , Rhodopsin/biosynthesis , cis-trans-Isomerases
11.
Invest Ophthalmol Vis Sci ; 46(11): 3988-98, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16249472

ABSTRACT

PURPOSE: Light-driven protein translocation is responsible for the dramatic redistribution of some proteins in vertebrate rod photoreceptors. In this study, the involvement of microtubules and microfilaments in the light-driven translocation of arrestin and transducin was investigated. METHODS: Pharmacologic reagents were applied to native and transgenic Xenopus tadpoles, to disrupt the microtubules (thiabendazole) and microfilaments (cytochalasin D and latrunculin B) of the rod photoreceptors. Quantitative confocal imaging was used to assess the impact of these treatments on arrestin and transducin translocation. A series of transgenic tadpoles expressing arrestin truncations were also created to identify portions of arrestin that enable arrestin to translocate. RESULTS: Application of cytochalasin D or latrunculin B to disrupt the microfilament organization selectively slowed only transducin movement from the inner to the outer segments. Perturbation of the microtubule cytoskeleton with thiabendazole slowed the translocation of both arrestin and transducin, but only in moving from the outer to the inner segments. Transgenic Xenopus expressing fusions of green fluorescent protein (GFP) with portions of arrestin implicates the C terminus of arrestin as an important portion of the molecule for promoting translocation. This C-terminal region can be used independently to promote translocation of GFP in response to light. CONCLUSIONS: The results show that disruption of the cytoskeletal network in rod photoreceptors has specific effects on the translocation of arrestin and transducin. These effects suggest that the light-driven translocation of visual proteins at least partially relies on an active motor-driven mechanism for complete movement of arrestin and transducin.


Subject(s)
Actin Cytoskeleton/physiology , Arrestin/metabolism , Light , Microtubules/physiology , Retinal Rod Photoreceptor Cells/metabolism , Transducin/metabolism , Actin Cytoskeleton/drug effects , Animals , Animals, Genetically Modified , Bridged Bicyclo Compounds, Heterocyclic/toxicity , Cytochalasin D/toxicity , Green Fluorescent Proteins/metabolism , Microscopy, Confocal , Microtubules/drug effects , Protein Transport/radiation effects , Recombinant Fusion Proteins/metabolism , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/radiation effects , Thiabendazole/toxicity , Thiazoles/toxicity , Thiazolidines , Xenopus laevis
12.
Mol Vis ; 11: 152-62, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15765048

ABSTRACT

PURPOSE: To report the phenotype and characterization of a new, naturally occurring mouse model of hereditary retinal degeneration (rd12). METHODS: The retinal phenotype of rd12 mice were studied using serial indirect ophthalmoscopy, fundus photography, electroretinography (ERG), genetic analysis including linkage studies and gene identification, immunohistochemistry, and biochemical analysis. RESULTS: Mice homozygous for the rd12 mutation showed small punctate white spots on fundus examination at 5 months of age. The retina in the rd12 homozygote had a normal appearance at the light microscopic level until 6 weeks of age when occasional voids appeared in the outer segments (OS) of the photoreceptor (PR) cells. The outer nuclear layer (ONL) appeared normal until 3 months of age though more obvious voids were detected in the OS. By 7 months of age, 6 to 8 layers of ONL remained in the mutant retina, and the OS were obviously shorter. The first sign of retinal degeneration was detected at the electron microscopic level around 3 weeks of age when occasional small lipid-like droplets were detected in the retinal pigment epithelium (RPE). By 3 months of age, much larger, lipid-like droplets accumulated in RPE cells accompanied by some OS degeneration. While the histology indicated a relatively slow retinal degeneration in the rd12 homozygous mutant mice, the rod ERG response was profoundly diminished even at 3 weeks of age. Genetic analysis showed that rd12 was an autosomal recessive mutation and mapped to mouse chromosome 3 closely linked to D3Mit19, a location known to be near the mouse Rpe65 gene. Sequence analysis showed that the mouse retinal degeneration is caused by a nonsense mutation in exon 3 of the Rpe65 gene, and the gene symbol for the rd12 mutation has been updated to Rpe65rd12 to reflect this. No RPE65 expression, 11-cis retinal, or rhodopsin could be detected in retinas from rd12 homozygotes, while retinyl esters were found to accumulate in the retinal pigment epithelium (RPE). CONCLUSIONS: Mutations in the retinal pigment epithelium gene encoding RPE65 cause an early onset autosomal recessive form of human retinitis pigmentosa, known as Leber congenital amaurosis (LCA), which results in blindness or severely impaired vision in children. A naturally arising mouse Rpe65 mutation provides a good model for studying the pathology of human RPE65 mutations and the effects of retinyl ester accumulation.


Subject(s)
Blindness/congenital , Codon, Nonsense , Disease Models, Animal , Eye Proteins/genetics , Retinal Degeneration/genetics , Animals , Blindness/metabolism , Blindness/pathology , Carrier Proteins , Electroretinography , Exons/genetics , Eye Proteins/metabolism , Female , Fluorescent Antibody Technique, Indirect , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Ophthalmoscopy , Phenotype , Photoreceptor Cells, Vertebrate/pathology , Pigment Epithelium of Eye/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinaldehyde/metabolism , Rhodopsin/metabolism , cis-trans-Isomerases
13.
J Mol Biol ; 343(5): 1439-50, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15491622

ABSTRACT

Rhodopsin, the pigment protein responsible for dim-light vision, is a G protein-coupled receptor that converts light absorption into the activation of a G protein, transducin, to initiate the visual response. We have crystallised detergent-solubilised bovine rhodopsin in the native form and after chemical modifications as needles 10-40 microm in cross-section. The crystals belong to the trigonal space group P3(1), with two molecules of rhodopsin per asymmetric unit, related by a non-crystallographic 2-fold axis parallel with the crystallographic screw axis along c (needle axis). The unit cell dimensions are a=103.8 A, c=76.6 A for native rhodopsin, but vary over a wide range after heavy atom derivatisation, with a between 101.5 A and 113.9 A, and c between 76.6 A and 79.2 A. Rhodopsin molecules are packed with the bundle of transmembrane helices tilted from the c-axis by about 100 degrees . The two molecules in the asymmetric unit form contacts along the entire length of their transmembrane helices 5 in an antiparallel orientation, and they are stacked along the needle axis according to the 3-fold screw symmetry. Hence hydrophobic contacts are prominent at protein interfaces both along and normal to the needle axis. The best crystals of native rhodopsin in this crystal form diffracted X-rays from a microfocused synchrotron source to 2.55 A maximum resolution. We describe steps taken to extend the diffraction limit from about 10 A to 2.6 A.


Subject(s)
Metals, Heavy/chemistry , Rhodopsin/chemistry , Animals , Cattle , Crystallization , Crystallography, X-Ray , Detergents , Metals, Heavy/metabolism , Protein Structure, Tertiary , Retina/chemistry , Retina/metabolism , Rhodopsin/analogs & derivatives , Rhodopsin/isolation & purification , Rhodopsin/metabolism , Spectrophotometry
14.
J Biol Chem ; 279(49): 51203-7, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15351781

ABSTRACT

Phosphorylation of activated G-protein-coupled receptors and the subsequent binding of arrestin mark major molecular events of homologous desensitization. In the visual system, interactions between arrestin and the phosphorylated rhodopsin are pivotal for proper termination of visual signals. By using high resolution proton nuclear magnetic resonance spectroscopy of the phosphorylated C terminus of rhodopsin, represented by a synthetic 7-phosphopolypeptide, we show that the arrestin-bound conformation is a well ordered helix-loop structure connected to rhodopsin via a flexible linker. In a model of the rhodopsin-arrestin complex, the phosphates point in the direction of arrestin and form a continuous negatively charged surface, which is stabilized by a number of positively charged lysine and arginine residues of arrestin. Opposite to the mostly extended structure of the unphosphorylated C-terminal domain of rhodopsin, the arrestin-bound C-terminal helix is a compact domain that occupies a central position between the cytoplasmic loops and occludes the key binding sites of transducin. In conjunction with other binding sites, the helix-loop structure provides a mechanism of shielding phosphates in the center of the rhodopsin-arrestin complex and appears critical in guiding arrestin for high affinity binding with rhodopsin.


Subject(s)
Arrestins/chemistry , Rhodopsin/chemistry , Animals , Arginine/chemistry , Binding Sites , Cattle , Cytoplasm/metabolism , Dose-Response Relationship, Drug , GTP-Binding Proteins/chemistry , Guanosine Triphosphate/chemistry , Lysine/chemistry , Magnetic Resonance Spectroscopy , Models, Molecular , Peptides/chemistry , Phosphorylation , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Serine/chemistry , Signal Transduction
15.
Mol Vis ; 10: 392-8, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15215746

ABSTRACT

PURPOSE: The binding of visual arrestin to phosphorylated, activated rhodopsin serves as a model for studying the inactivation process of a large class of G-protein coupled receptor systems. In this study, we combine the use of insertional mutagenesis, fluorescence labeling, and scanning alanine mutagenesis to identify the surface of interaction between arrestin and rhodopsin. METHODS: The ten amino acid myc tag (EQKLISEEDL) was inserted in eleven loop structures that connect betastrands and the tagged arrestins were heterologously expressed in yeast. Binding competition assays were performed with these proteins, using an anti-myc monoclonal antibody. Site specific cysteines were also substituted in selected loop structures in arrestin. These cysteines were labeled with a fluorescent reporter to assess the proximity of the introduced cysteine with rhodopsin in the bound complex. RESULTS: Competitive inhibition of arrestin binding to light activated, phosphorylated rhodopsin with an anti-myc antibody showed that all competitive sites lay along a single surface encompassing the N- and C-terminal domains. Fluorescence labeling of these loop structures and subsequent interaction with rhodopsin indicates close apposition of loops 68-78 and 248-253 to rhodopsin in the receptor bound state. Scanning mutagenesis of loop 248-253 implicates Ser-251 and/or Ser-252 as a potential interaction point with rhodopsin. CONCLUSIONS: Our results clearly suggest a surface of arrestin to which rhodopsin binds upon light activation and phosphorylation. This surface encompasses elements from both the N- and C-terminal domains of arrestin.


Subject(s)
Arrestin/metabolism , Rhodopsin/metabolism , Arrestin/chemistry , Binding, Competitive , Mutagenesis, Insertional , Peptide Fragments/metabolism , Phosphorylation , Protein Binding , Protein Conformation
16.
FEBS Lett ; 564(3): 307-11, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-15111114

ABSTRACT

Visual arrestin binds to the phosphorylated carboxy-terminal region of rhodopsin to block interactions with transducin and terminate signaling in the rod photoreceptor cells. A synthetic seven-phospho-peptide from the C-terminal region of rhodopsin, Rh(330-348), has been shown to bind arrestin and mimic inhibition of signal transduction. In this study, we examine conformational changes in this synthetic peptide upon binding to arrestin by high-resolution proton nuclear magnetic resonance (NMR). We show that the peptide is completely disordered in solution, but becomes structured upon binding to arrestin. A control, unphosphorylated peptide that fails to bind to arrestin remains highly disordered. Specific NMR distance constraints are used to model the arrestin-bound conformation. The models suggest that the phosphorylated carboxy-terminal region of rhodopsin, Rh(330-348), undergoes significant conformational changes and becomes structured upon binding to arrestin.


Subject(s)
Arrestin/chemistry , Arrestin/metabolism , Protein Conformation , Rhodopsin/chemistry , Rhodopsin/metabolism , Animals , Cattle , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Protein Binding , Signal Transduction
17.
J Biol Chem ; 279(16): 16278-84, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-14769795

ABSTRACT

The clinically common mutant opsin P23H, associated with autosomal dominant retinitis pigmentosa, yields low levels of rhodopsin when retinal is added following induction of the protein in stably transfected HEK-293 cells. We previously showed that P23H rhodopsin levels could be increased by providing a 7-membered ring, locked analog of 11-cis-retinal during expression of P23H opsin in vivo. Here we demonstrate that the mutant opsin is effectively rescued by 9- or 11-cis-retinal, the native chromophore. When retinal was added during expression, P23H rhodopsin levels were 5-fold (9-cis) and 6-fold (11-cis) higher than when retinal was added after opsin was expressed and cells were harvested. Levels of P23H opsin were increased approximately 3.5-fold with both compounds, but wild-type protein levels were only slightly increased. Addition of retinal during induction promoted the Golgi-specific glycosylation of P23H opsin and transport of the protein to the cell surface. P23H rhodopsins containing 9- or 11-cis-retinal had blue-shifted absorption maxima and altered photo-bleaching properties compared with the corresponding wild-type proteins. Significantly, P23H rhodopsins were more thermally unstable than the wild-type proteins and more rapidly bleached by hydroxylamine in the dark. We suggest that P23H opsin is similarly unstable and that retinal binds and stabilizes the protein early in its biogenesis to promote its cellular folding and trafficking. The implications of this study for treating retinitis pigmentosa and other protein conformational disorders are discussed.


Subject(s)
Rod Opsins/chemistry , Humans , Mutation , Protein Conformation , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinoids/chemistry , Retinoids/metabolism , Rhodopsin/chemistry , Rhodopsin/metabolism , Rod Opsins/genetics , Rod Opsins/metabolism , Temperature
18.
Exp Eye Res ; 76(5): 553-63, 2003 May.
Article in English | MEDLINE | ID: mdl-12697419

ABSTRACT

Subcellular translocation of phototransduction proteins in response to light has previously been detected by immunocytochemistry. This movement is consistent with the hypothesis that migration is part of a basic cellular mechanism regulating photoreceptor sensitivity. In order to monitor the putative migration of arrestin in response to light, we expressed a functional fusion between the signal transduction protein arrestin and green fluorescent protein (GFP) in rod photoreceptors of transgenic Xenopus laevis. In addition to confirming reports that arrestin is translocated, this alternative approach generated unique observations, raising new questions regarding the nature and time scale of migration. Confocal fluorescence microscopy was performed on fixed frozen retinal sections from tadpoles exposed to three different lighting conditions. A consistent pattern of localization emerged in each case. During early light exposure, arrestin-GFP levels diminished in the inner segments (ISs) and simultaneously increased in the outer segments (OSs), initially at the base and eventually at the distal tips as time progressed. Arrestin-GFP reached the distal tips of the photoreceptors by 45-75 min at which time the ratio of arrestin-GFP fluorescence in the OSs compared to the ISs was maximal. When dark-adaptation was initiated after 45 min of light exposure, arrestin-GFP rapidly re-localized to the ISs and axoneme within 30 min. Curiously, prolonged periods of light exposure also resulted in re-localization of arrestin-GFP. Between 150 and 240 min of light adaptation the arrestin-GFP in the ROS gradually declined until the pattern of arrestin-GFP localization was indistinguishable from that of dark-adapted photoreceptors. This distribution pattern was observed over a wide range of lighting intensity (25-2700 lux). Immunocytochemical analysis of arrestin in wild-type Xenopus retinas gave similar results.


Subject(s)
Adaptation, Ocular/physiology , Arrestin/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Animals, Genetically Modified , Dark Adaptation/physiology , Green Fluorescent Proteins , Light , Luminescent Proteins , Microscopy, Confocal , Photic Stimulation , Recombinant Fusion Proteins/metabolism , Rod Cell Outer Segment/metabolism , Xenopus laevis
19.
J Clin Invest ; 111(8): 1171-80, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12697736

ABSTRACT

Experimental autoimmune uveitis (EAU) is a disease of the neural retina induced by immunization with retinal antigens, such as interphotoreceptor retinoid-binding protein (IRBP) and arrestin (retinal soluble antigen, S-Ag). EAU serves as a model for human autoimmune uveitic diseases associated with major histocompatibility complex (HLA) genes, in which patients exhibit immunological responses to retinal antigens. Here we report the development of a humanized EAU model in HLA transgenic (TG) mice. HLA-DR3, -DR4, -DQ6, and -DQ8 TG mice were susceptible to IRBP-induced EAU. Importantly, HLA-DR3 TG mice developed severe EAU with S-Ag, to which wild-type mice are highly resistant. Lymphocyte proliferation was blocked by anti-HLA antibodies, confirming that antigen is functionally presented by the human MHC molecules. Disease could be transferred by immune cells with a Th1-like cytokine profile. Antigen-specific T cell repertoire, as manifested by responses to overlapping peptides derived from S-Ag or IRBP, differed from that of wild-type mice. Interestingly, DR3 TG mice, but not wild-type mice, recognized an immunodominant S-Ag epitope between residues 291 and 310 that overlaps with a region of S-Ag recognized by uveitis patients. Thus, EAU in HLA TG mice offers a new model of uveitis that should represent human disease more faithfully than currently existing models.


Subject(s)
Autoimmune Diseases/etiology , Eye Proteins , Histocompatibility Antigens Class II/physiology , Uveitis/etiology , Amino Acid Sequence , Animals , Antigen Presentation , Arrestin/immunology , Disease Models, Animal , Epitopes, T-Lymphocyte , HLA-DR3 Antigen/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Retinol-Binding Proteins/immunology
20.
Eur J Biochem ; 269(15): 3801-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12153577

ABSTRACT

Visual arrestin is converted from a 'basal' state to an 'activated' state by interaction with the phosphorylated C-terminus of photoactivated rhodopsin (R*), but the conformational changes in arrestin that lead to activation are unknown. Small-angle X-ray scattering (SAXS) was used to investigate the solution structure of arrestin and characterize changes attendant upon activation. Wild-type arrestin forms dimers with a dissociation constant of 60 micro m. Small conformational changes, consistent with local movements of loops or the mobile N- or C-termini of arrestin, were observed in the presence of a phosphopeptide corresponding to the C-terminus of rhodopsin, and with an R175Q mutant. Because both the phosphopeptide and the R175Q mutation promote binding to unphosphorylated R*, we conclude that arrestin is activated by subtle conformational changes. Most of the arrestin will be in a dimeric state in vivo. Using the arrestin structure as a guide [Hirsch, J.A., Schubert, C., Gurevich, V.V. & Sigler, P.B. (1999) Cell 97, 257-269], we have identified a model for the arrestin dimer that is consistent with our SAXS data. In this model, dimerization is mediated by the C-terminal domain of arrestin, leaving the N-terminal domains free for interaction with phosphorylated R*.


Subject(s)
Arrestin/chemistry , Amino Acid Sequence , Arrestin/genetics , Arrestin/metabolism , Dimerization , Models, Molecular , Molecular Sequence Data , Mutation , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphorylation , Protein Conformation , Rhodopsin/chemistry , Rhodopsin/genetics , Rhodopsin/metabolism , Scattering, Radiation , Solutions , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...