Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 34(40): 13336-48, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25274813

ABSTRACT

Retinitis pigmentosa (RP) is an inherited neurodegenerative disease involving progressive vision loss, and is often linked to mutations in the rhodopsin gene. Mutations that abolish N-terminal glycosylation of rhodopsin (T4K and T17M) cause sector RP in which the inferior retina preferentially degenerates, possibly due to greater light exposure of this region. Transgenic animal models expressing rhodopsin glycosylation mutants also exhibit light exacerbated retinal degeneration (RD). In this study, we used transgenic Xenopus laevis to investigate the pathogenic mechanism connecting light exposure and RD in photoreceptors expressing T4K or T17M rhodopsin. We demonstrate that increasing the thermal stability of these rhodopsins via a novel disulfide bond resulted in significantly less RD. Furthermore, T4K or T17M rhodopsins that were constitutively inactive (due to lack of the chromophore-binding site or dietary deprivation of the chromophore precursor vitamin A) induced less toxicity. In contrast, variants in the active conformation accumulated in the ER and caused RD even in the absence of light. In vitro, T4K and T17M rhodopsins showed reduced ability to regenerate pigment after light exposure. Finally, although multiple amino acid substitutions of T4 abolished glycosylation at N2 but were not toxic, similar substitutions of T17 were not tolerated, suggesting that the carbohydrate moiety at N15 is critical for cell viability. Our results identify a novel pathogenic mechanism in which the glycosylation-deficient rhodopsins are destabilized by light activation. These results have important implications for proposed RP therapies, such as vitamin A supplementation, which may be ineffective or even detrimental for certain RP genotypes.


Subject(s)
Light , Mutation/genetics , Retinal Degeneration/etiology , Retinitis Pigmentosa , Rhodopsin/genetics , Rod Cell Outer Segment/pathology , Analysis of Variance , Animals , Animals, Genetically Modified , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Humans , Microscopy, Confocal , Retinal Degeneration/diet therapy , Retinitis Pigmentosa/complications , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Statistics, Nonparametric , Transfection , Vitamin A/administration & dosage , Vitamin A/metabolism , Wheat Germ Agglutinins/metabolism , Xenopus laevis
2.
Proc Natl Acad Sci U S A ; 107(13): 5961-6, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20231467

ABSTRACT

The P23H mutation within the rhodopsin gene (RHO) causes rhodopsin misfolding, endoplasmic reticulum (ER) stress, and activates the unfolded protein response (UPR), leading to rod photoreceptor degeneration and autosomal dominant retinitis pigmentosa (ADRP). Grp78/BiP is an ER-localized chaperone that is induced by UPR signaling in response to ER stress. We have previously demonstrated that BiP mRNA levels are selectively reduced in animal models of ADRP arising from P23H rhodopsin expression at ages that precede photoreceptor degeneration. We have now overexpressed BiP to test the hypothesis that this chaperone promotes the trafficking of P23H rhodopsin to the cell membrane, reprograms the UPR favoring the survival of photoreceptors, blocks apoptosis, and, ultimately, preserves vision in ADRP rats. In cell culture, increasing levels of BiP had no impact on the localization of P23H rhodopsin. However, BiP overexpression alleviated ER stress by reducing levels of cleaved pATF6 protein, phosphorylated eIF2alpha and the proapoptotic protein CHOP. In P23H rats, photoreceptor levels of cleaved ATF6, pEIF2alpha, CHOP, and caspase-7 were much higher than those of wild-type rats. Subretinal delivery of AAV5 expressing BiP to transgenic rats led to reduction in CHOP and photoreceptor apoptosis and to a sustained increase in electroretinogram amplitudes. We detected complexes between BiP, caspase-12, and the BH3-only protein BiK that may contribute to the antiapoptotic activity of BiP. Thus, the preservation of photoreceptor function resulting from elevated levels of BiP is due to suppression of apoptosis rather than to a promotion of rhodopsin folding.


Subject(s)
Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism , Vision, Ocular/genetics , Vision, Ocular/physiology , Amino Acid Substitution , Animals , Apoptosis , Base Sequence , Dependovirus/genetics , Disease Models, Animal , Electroretinography , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , HeLa Cells , Humans , Mice , Models, Biological , Multiprotein Complexes , Mutation, Missense , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retina/pathology , Retina/physiopathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/physiopathology , Retinitis Pigmentosa/therapy , Rhodopsin/chemistry , Stress, Physiological , Transcription Factor CHOP/metabolism , Transfection , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology
3.
J Biol Chem ; 284(48): 33333-42, 2009 Nov 27.
Article in English | MEDLINE | ID: mdl-19801547

ABSTRACT

The lectin chaperone calnexin (Cnx) is important for quality control of glycoproteins, and the chances of correct folding of a protein increase the longer the protein interacts with Cnx. Mutations in glycoproteins increase their association with Cnx, and these mutant proteins are retained in the endoplasmic reticulum. However, until now, the increased interaction with Cnx was not known to increase the folding of mutant glycoproteins. Because many human diseases result from glycoprotein misfolding, a Cnx-assisted folding of mutant glycoproteins could be beneficial. Mutations of rhodopsin, the glycoprotein pigment of rod photoreceptors, cause misfolding resulting in retinitis pigmentosa. Despite the critical role of Cnx in glycoprotein folding, surprisingly little is known about its interaction with rhodopsin or whether this interaction could be modulated to increase the folding of mutant rhodopsin. Here, we demonstrate that Cnx preferentially associates with misfolded mutant opsins associated with retinitis pigmentosa. Furthermore, the overexpression of Cnx leads to an increased accumulation of misfolded P23H opsin but not the correctly folded protein. Finally, we demonstrate that increased levels of Cnx in the presence of the pharmacological chaperone 11-cis-retinal increase the folding efficiency and result in an increase in correct folding of mutant rhodopsin. These results demonstrate that misfolded rather than correctly folded rhodopsin is a substrate for Cnx and that the interaction between Cnx and mutant, misfolded rhodopsin, can be targeted to increase the yield of folded mutant protein.


Subject(s)
Calnexin/metabolism , Mutation , Retinaldehyde/metabolism , Rhodopsin/metabolism , Animals , Blotting, Western , Calnexin/pharmacology , Cell Line , Endoplasmic Reticulum/metabolism , Humans , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding/drug effects , Protein Folding/drug effects , Retinaldehyde/pharmacology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Rhodopsin/chemistry , Rhodopsin/genetics
4.
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
5.
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
6.
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
7.
Trends Biochem Sci ; 29(12): 648-55, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15544951

ABSTRACT

Integral membrane proteins (IMPs) are essential components of the plasma and organellar membranes of the eukaryotic cell. Non-native IMPs, which can arise as a result of mutations, errors during biosynthesis or cellular stress, can disrupt these membranes and potentially lead to cell death. To protect against this outcome, the cell possesses quality control (QC) systems that detect and dispose of non-native IMPs from cellular membranes. Recent studies suggest that recognition of non-native IMPs by the QC machinery is correlated with the thermodynamic stability of these proteins. Consistent with this, small molecules known as chemical and pharmacological chaperones have been identified that stabilize non-native IMPs and enable them to evade QC. These findings have far-reaching implications for treating human diseases caused by defective IMPs.


Subject(s)
Cell Membrane/physiology , Membrane Proteins/physiology , Models, Biological , Molecular Chaperones , Protein Folding , Quality Control
8.
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
9.
J Biol Chem ; 278(16): 14442-14450, 2003 Apr 18.
Article in English | MEDLINE | ID: mdl-12566452

ABSTRACT

Protein conformational disorders, which include certain types of retinitis pigmentosa, are a set of inherited human diseases in which mutant proteins are misfolded and often aggregated. Many opsin mutants associated with retinitis pigmentosa, the most common being P23H, are misfolded and retained within the cell. Here, we describe a pharmacological chaperone, 11-cis-7-ring retinal, that quantitatively induces the in vivo folding of P23H-opsin. The rescued protein forms pigment, acquires mature glycosylation, and is transported to the cell surface. Additionally, we determined the temperature stability of the rescued protein as well as the reactivity of the retinal-opsin Schiff base to hydroxylamine. Our study unveils novel properties of P23H-opsin and its interaction with the chromophore. These properties suggest that 11-cis-7-ring retinal may be a useful therapeutic agent for the rescue of P23H-opsin and the prevention of retinal degeneration.


Subject(s)
Retinitis Pigmentosa/genetics , Rod Opsins/chemistry , Rod Opsins/genetics , Amino Acid Sequence , Cell Line , Cell Membrane/metabolism , Chromatography, High Pressure Liquid , Detergents/pharmacology , Electrophoresis, Polyacrylamide Gel , Genes, Dominant , Glycosylation , Humans , Hydrogen-Ion Concentration , Hydroxylamines/metabolism , Immunoblotting , Immunohistochemistry , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Mutation , Protein Conformation , Protein Folding , Protein Structure, Secondary , Temperature , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...