Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Exp Eye Res ; 239: 109760, 2024 Feb.
Article En | MEDLINE | ID: mdl-38158174

Rod and cone photoreceptors are named for the distinct morphologies of their outer segment organelles, which are either cylindrical or conical, respectively. The morphologies of the stacked disks that comprise the rod and cone outer segments also differ: rod disks are completely sealed and are discontinuous from the plasma membrane, while cone disks remain partially open to the extracellular space. These morphological differences between photoreceptor types are more prominent in non-mammalian vertebrates, whose cones typically possess a greater proportion of open disks and are more tapered in shape. In mammals, the tetraspanin prph2 generates and maintains the highly curved disk rim regions by forming extended oligomeric structures with itself and a structurally similar paralog, rom1. Here we determined that in addition to these two proteins, there is a third Prph2 family paralog in most non-mammalian vertebrate species, including X. laevis: Glycoprotein 2-like protein or "Gp2l". A survey of multiple genome databases revealed a single invertebrate Prph2 'pro-ortholog' in Amphioxus, several echinoderms and in a diversity of protostomes indicating an ancient divergence from other tetraspanins. Based on phylogenetic analysis, duplication of the vertebrate predecessor likely gave rise to the Gp2l and Prph2/Rom1 clades, with a further duplication distinguishing the Prph2 and Rom1 clades. Mammals have lost Gp2l and their Rom1 has undergone a period of accelerated evolution such that it has lost several features that are retained in non-mammalian vertebrate Rom1. Specifically, Prph2, Gp2l and non-mammalian Rom1 encode proteins with consensus N-linked glycosylation and outer segment localization signals; mammalian rom1 lacks these motifs. We determined that X. laevis gp2l is expressed exclusively in cones and green rods, while X. laevis rom1 is expressed exclusively in rods, and prph2 is present in both rods and cones. The presence of three Prph2-related genes with distinct expression patterns as well as the rapid evolution of mammalian Rom1, may contribute to the more pronounced differences in morphology between rod and cone outer segments and rod and cone disks observed in non-mammalian versus mammalian vertebrates.


Retinal Degeneration , Animals , Gene Duplication , Mammals , Peripherins/genetics , Peripherins/metabolism , Phylogeny , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Tetraspanins/genetics , Xenopus laevis/genetics , Xenopus laevis/metabolism
2.
Mol Ther Methods Clin Dev ; 17: 478-490, 2020 Jun 12.
Article En | MEDLINE | ID: mdl-32258211

Aniridia is a rare eye disorder, which is caused by mutations in the paired box 6 (PAX6) gene and results in vision loss due to the lack of a long-term vision-saving therapy. One potential approach to treating aniridia is targeted CRISPR-based genome editing. To enable the Pax6 small eye (Sey) mouse model of aniridia, which carries the same mutation found in patients, for preclinical testing of CRISPR-based therapeutic approaches, we endogenously tagged the Sey allele, allowing for the differential detection of protein from each allele. We optimized a correction strategy in vitro then tested it in vivo in the germline of our new mouse to validate the causality of the Sey mutation. The genomic manipulations were analyzed by PCR, as well as by Sanger and next-generation sequencing. The mice were studied by slit lamp imaging, immunohistochemistry, and western blot analyses. We successfully achieved both in vitro and in vivo germline correction of the Sey mutation, with the former resulting in an average 34.8% ± 4.6% SD correction, and the latter in restoration of 3xFLAG-tagged PAX6 expression and normal eyes. Hence, in this study we have created a novel mouse model for aniridia, demonstrated that germline correction of the Sey mutation alone rescues the mutant phenotype, and developed an allele-distinguishing CRISPR-based strategy for aniridia.

3.
Invest Ophthalmol Vis Sci ; 60(4): 933-943, 2019 03 01.
Article En | MEDLINE | ID: mdl-30840038

Purpose: The rhodopsin mutation P23H is responsible for a significant portion of autosomal-dominant retinitis pigmentosa, a disorder characterized by rod photoreceptor death. The mechanisms of toxicity remain unclear; previous studies implicate destabilization of P23H rhodopsin during light exposure, causing decreased endoplasmic reticulum (ER) exit and ER stress responses. Here, we probed phototransduction in Xenopus laevis rods expressing bovine P23H rhodopsin, in which retinal degeneration is inducible by light exposure, in order to examine early physiological changes that occur during retinal degeneration. Methods: We recorded single-cell and whole-retina responses to light stimuli using electrophysiology. Moreover, we monitored morphologic changes in rods after different periods of light exposure. Results: Initially, P23H rods had almost normal photoresponses, but following a brief light exposure varying from 4 to 32 photoisomerizations per disc, photoresponses became irreversibly prolonged. In intact retinas, rods began to shed OS fragments after a rod-saturating exposure of 12 minutes, corresponding to approximately 10 to 100 times more photoisomerizations. Conclusions: Our results indicate that in P23H rods light-induced degeneration occurs in at least two stages, the first involving impairment of phototransduction and the second involving initiation of morphologic changes.


Animals, Genetically Modified , Retinitis Pigmentosa/physiopathology , Rhodopsin/genetics , Rod Cell Outer Segment/physiology , Vision, Ocular/physiology , Animals , Dark Adaptation/physiology , Disease Models, Animal , Electrophysiological Phenomena , Electroretinography , Female , Male , Microscopy, Confocal , Photic Stimulation , Retinitis Pigmentosa/genetics , Xenopus laevis
4.
Methods Mol Biol ; 1834: 193-207, 2019.
Article En | MEDLINE | ID: mdl-30324446

Xenopus laevis have proven to be a useful system for rapid generation and analysis of transgenic models of human retinal disease. However, experimental approaches in this system were limited by lack of a robust knockdown or knockout technology. Here we describe a protocol for generation of Cas9-edited X. laevis embryos. The technique introduces point mutations into the genome of X. laevis resulting in in-frame and out-of-frame insertions and deletions that allow modeling of both dominant and recessive human diseases and efficiently generates gene knockdown and knockout. Our techniques can produce high-frequency gene editing in X. laevis, permitting analysis in the F0 generation.


CRISPR-Cas Systems , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Animals , Disease Models, Animal , Fluorescent Antibody Technique , Gene Editing , Gene Expression , Gene Knockout Techniques , Genes, Reporter , Humans , Mice , Phenotype , RNA, Guide, Kinetoplastida , Retinal Degeneration/pathology , Xenopus laevis
5.
J Cell Sci ; 131(24)2018 12 10.
Article En | MEDLINE | ID: mdl-30404838

The Arf4-rhodopsin complex (mediated by the VxPx motif in rhodopsin) initiates expansion of vertebrate rod photoreceptor cilia-derived light-sensing organelles through stepwise assembly of a conserved trafficking network. Here, we examine its role in the sorting of VAMP7 (also known as TI-VAMP) - an R-SNARE possessing a regulatory longin domain (LD) - into rhodopsin transport carriers (RTCs). During RTC formation and trafficking, VAMP7 colocalizes with the ciliary cargo rhodopsin and interacts with the Rab11-Rabin8-Rab8 trafficking module. Rab11 and Rab8 bind the VAMP7 LD, whereas Rabin8 (also known as RAB3IP) interacts with the SNARE domain. The Arf/Rab11 effector FIP3 (also known as RAB11FIP3) regulates VAMP7 access to Rab11. At the ciliary base, VAMP7 forms a complex with the cognate SNAREs syntaxin 3 and SNAP-25. When expressed in transgenic animals, a GFP-VAMP7ΔLD fusion protein and a Y45E phosphomimetic mutant colocalize with endogenous VAMP7. The GFP-VAMP7-R150E mutant displays considerable localization defects that imply an important role of the R-SNARE motif in intracellular trafficking, rather than cognate SNARE pairing. Our study defines the link between VAMP7 and the ciliary targeting nexus that is conserved across diverse cell types, and contributes to general understanding of how functional Arf and Rab networks assemble SNAREs in membrane trafficking.


ADP-Ribosylation Factors/metabolism , Cilia/metabolism , Membrane Fusion/physiology , SNARE Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Organelles/metabolism , Protein Transport/physiology , R-SNARE Proteins/metabolism , Rhodopsin/metabolism
6.
Sci Rep ; 7(1): 6920, 2017 07 31.
Article En | MEDLINE | ID: mdl-28761125

The utility of Xenopus laevis, a common research subject for developmental biology, retinal physiology, cell biology, and other investigations, has been limited by lack of a robust gene knockout or knock-down technology. Here we describe manipulation of the X. laevis genome using CRISPR/Cas9 to model the human disorder retinitis pigmentosa, and to introduce point mutations or exogenous DNA sequences. We introduced and characterized in-frame and out-of-frame insertions and deletions in three genes encoding rhodopsin by co-injection of Cas9 mRNA, eGFP mRNA, and single guide RNAs into fertilized eggs. Deletions were characterized by direct sequencing and cloning; phenotypes were assessed by assays of rod opsin in retinal extracts, and confocal microscopy of cryosectioned and immunolabeled contralateral eyes. We obtained germline transmission of editing to F1 offspring. In-frame deletions frequently caused dominant retinal degeneration associated with rhodopsin biosynthesis defects, while frameshift phenotypes were consistent with knockout. We inserted eGFP or point mutations into rhodopsin genes by co-injection of repair fragments with homology to the Cas9 target sites. Our techniques can produce high frequency gene editing in X. laevis, permitting analysis in the F0 generation, and advancing the utility of X. laevis as a subject for biological research and disease modeling.


Disease Models, Animal , Gene Editing/methods , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , Animals , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Female , Genes, Dominant , Genes, Recessive , Green Fluorescent Proteins/genetics , Humans , Male , Phenotype , Point Mutation , RNA, Guide, Kinetoplastida/genetics , Retinitis Pigmentosa/pathology , Sequence Deletion , Xenopus Proteins/genetics , Xenopus laevis/embryology
7.
J Neurosci ; 37(4): 1039-1054, 2017 01 25.
Article En | MEDLINE | ID: mdl-28490005

Retinitis pigmentosa (RP) is an inherited retinal degeneration (RD) that leads to blindness for which no treatment is available. RP is frequently caused by mutations in Rhodopsin; in some animal models, RD is exacerbated by light. Valproic acid (VPA) is a proposed treatment for RP and other neurodegenerative disorders, with a phase II trial for RP under way. However, the therapeutic mechanism is unclear, with minimal research supporting its use in RP. We investigated the effects of VPA on Xenopus laevis models of RP expressing human P23H, T17M, T4K, and Q344ter rhodopsins, which are associated with RP in humans. VPA ameliorated RD associated with P23H rhodopsin and promoted clearing of mutant rhodopsin from photoreceptors. The effect was equal to that of dark rearing, with no additive effect observed. Rescue of visual function was confirmed by electroretinography. In contrast, VPA exacerbated RD caused by T17M rhodopsin in light, but had no effect in darkness. Effects in T4K and Q344ter rhodopsin models were also negative. These effects of VPA were paralleled by treatment with three additional histone deacetylase (HDAC) inhibitors, but not other antipsychotics, chemical chaperones, or VPA structural analogues. In WT retinas, VPA treatment increased histone H3 acetylation. In addition, electron microscopy showed increased autophagosomes in rod inner segments with HDAC inhibitor (HDACi) treatment, potentially linking the therapeutic effects in P23H rhodopsin animals and negative effects in other models with autophagy. Our results suggest that the success or failure of VPA treatment is dependent on genotype and that HDACi treatment is contraindicated for some RP cases.SIGNIFICANCE STATEMENT Retinitis pigmentosa (RP) is an inherited, degenerative retinal disease that leads to blindness for which no therapy is available. We determined that valproic acid (VPA), currently undergoing a phase II trial for RP, has both beneficial and detrimental effects in animal models of RP depending on the underlying disease mechanism and that both effects are due to histone deacetylase (HDAC) inhibition possibly linked to autophagy regulation. Off-label use of VPA and other HDAC inhibitors for the treatment of RP should be limited to the research setting until this effect is understood and can be predicted. Our study suggests that, unless genotype is accounted for, clinical trials for RP treatments may give negative results due to multiple disease mechanisms with differential responses to therapeutic interventions.


Histone Deacetylase Inhibitors/therapeutic use , Retinitis Pigmentosa/drug therapy , Valproic Acid/therapeutic use , Animals , Autophagosomes/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Photoreceptor Cells/drug effects , Photoreceptor Cells/metabolism , Photoreceptor Cells/ultrastructure , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Rhodopsin/genetics , Valproic Acid/pharmacology , Xenopus laevis
8.
FASEB J ; 29(12): 4866-80, 2015 Dec.
Article En | MEDLINE | ID: mdl-26229057

In Caenorhabditis elegans, homodimeric [kinesin family (KIF) 17, osmotic avoidance abnormal-3 (OSM-3)] and heterotrimeric (KIF3) kinesin-2 motors are required to establish sensory cilia by intraflagellar transport (IFT) where KIF3 and KIF17 cooperate to build the axoneme core and KIF17 builds the distal segments. However, the function of KIF17 in vertebrates is unresolved. We expressed full-length and motorless KIF17 constructs in mouse rod photoreceptors using adeno-associated virus in Xenopus laevis rod photoreceptors using a transgene and in ciliated IMCD3 cells. We found that tagged KIF17 localized along the rod outer segment axoneme when expressed in mouse and X. laevis photoreceptors, whereas KIF3A was restricted to the proximal axoneme. Motorless KIF3A and KIF17 mutants caused photoreceptor degeneration, likely through dominant negative effects on IFT. KIF17 mutant lacking the motor domain translocated to nuclei after exposure of a C-terminal nuclear localization signal. Germ-line deletion of Kif17 in mouse did not affect photoreceptor function. A rod-specific Kif3/Kif17 double knockout mouse demonstrated that KIF17 and KIF3 do not act synergistically and did not prevent rhodopsin trafficking to rod outer segments. In summary, the nematode model of KIF3/KIF17 cooperation apparently does not apply to mouse photoreceptors in which the photosensory cilium is built exclusively by KIF3.


Kinesins/physiology , Photoreceptor Cells, Vertebrate/physiology , Amino Acid Sequence , Animals , HEK293 Cells , Humans , Kinesins/chemistry , Kinesins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , NIH 3T3 Cells , Protein Transport , Rhodopsin/metabolism , Xenopus laevis
9.
Exp Eye Res ; 136: 86-90, 2015 Jul.
Article En | MEDLINE | ID: mdl-26008144

Transmission electron microscopy is the gold standard for examination of photoreceptor outer segment morphology and photoreceptor outer segment abnormalities in transgenic animal models of retinal disease. Small vertebrates such as zebrafish and Xenopus laevis tadpoles have been used to generate retinal disease models and to study outer segment processes such as protein trafficking, and their breeding capabilities facilitate experiments involving large numbers of animals and conditions. However, electron microscopy processing and analysis of these very small eyes can be challenging. Here we present a methodology that facilitates processing of X. laevis tadpole eyes for electron microscopy by introducing an intermediate cryosectioning step. This method reproducibly provides a well-oriented tissue block that can be sectioned with minimal effort by a non-expert, and also allows retroactive analysis of samples collected on slides for light microscopy.


Cryoultramicrotomy/methods , Retina/ultrastructure , Xenopus laevis , Animals , Histocytological Preparation Techniques , Microscopy, Electron, Transmission , Tissue Embedding , Tissue Fixation/methods
10.
J Neurosci ; 34(40): 13336-48, 2014 Oct 01.
Article En | MEDLINE | ID: mdl-25274813

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.


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
11.
Invest Ophthalmol Vis Sci ; 55(6): 3669-80, 2014 May 15.
Article En | MEDLINE | ID: mdl-24833735

PURPOSE: Autosomal dominant Stargardt macular dystrophy caused by mutations in the Elongation of Very Long Chain fatty acids (ELOVL4) gene results in macular degeneration, leading to early childhood blindness. Transgenic mice and pigs expressing mutant ELOVL4 develop progressive photoreceptor degeneration. The mechanism by which these mutations cause macular degeneration remains unclear, but have been hypothesized to involve the loss of an ER-retention dilysine motif located in the extreme C-terminus. Dominant negative mechanisms and reduction in retinal polyunsaturated fatty acids also have been suggested. To understand the molecular mechanisms involved in disease progression in vivo, we addressed the hypothesis that the disease-linked C-terminal truncation mutant of ELOVL4 exerts a dominant negative effect on wild-type (WT) ELOVL4, altering its subcellular localization and function, which subsequently induces retinal degeneration and loss of vision. METHODS: We generated transgenic Xenopus laevis that overexpress HA-tagged murine ELOVL4 variants in rod photoreceptors. RESULTS: Tagged or untagged WT ELOVL4 localized primarily to inner segments. However, the mutant protein lacking the dilysine motif was mislocalized to post-Golgi compartments and outer segment disks. Coexpression of mutant and WT ELOVL4 in rods did not result in mislocalization of the WT protein to outer segments or in the formation of aggregates. Full-length HA-tagged ELOVL4 lacking the dilysine motif (K308R/K310R) necessary for targeting the WT ELOVL4 protein to the endoplasmic reticulum was similarly mislocalized to outer segments. CONCLUSIONS: We propose that expression and outer segment mislocalization of the disease-linked 5-base-pair deletion mutant ELOVL4 protein alters photoreceptor structure and function, which subsequently results in retinal degeneration, and suggest three possible mechanisms by which mutant ELOVL4 may induce retinal degeneration in STGD3.


DNA/genetics , Eye Proteins/genetics , Macular Degeneration/congenital , Membrane Proteins/genetics , Mutation , Rod Cell Outer Segment/metabolism , Animals , DNA Mutational Analysis , Disease Models, Animal , Eye Proteins/metabolism , Immunohistochemistry , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Rod Cell Outer Segment/pathology , Xenopus laevis/genetics
12.
Methods Mol Biol ; 935: 113-25, 2013.
Article En | MEDLINE | ID: mdl-23150364

Transgenic models are invaluable tools for researching retinal degenerative disease mechanisms. However, they are time-consuming and expensive to generate and maintain. We have developed an alternative to transgenic rodent models of retinal degeneration using transgenic Xenopus laevis. We have optimized this system to allow rapid analysis of transgene effects in primary transgenic animals, thereby providing an alternative to establishing transgenic lines, and simultaneously allowing rigorous comparisons between the effects of different transgenes.


Animals, Genetically Modified/genetics , Disease Models, Animal , Retinal Degeneration/genetics , Xenopus laevis/genetics , Animals , Humans , Transgenes
13.
J Neurosci ; 32(6): 2121-8, 2012 Feb 08.
Article En | MEDLINE | ID: mdl-22323724

This study evaluated the capacity of Xenopus laevis retina to regenerate photoreceptor cells after cyclic light-mediated acute rod photoreceptor degeneration in a transgenic P23H mutant rhodopsin model of retinits pigmentosa. After discontinuation of cyclic light exposure, we monitored histologic progression of retinal regeneration over a 3 week recovery period. To assess their metabolomic states, contralateral eyes were processed for computational molecular phenotyping. We found that retinal degeneration in the P23H rhodopsin mutation could be partially reversed, with regeneration of rod photoreceptors recovering normal morphology (including full-length rod outer segments) by the end of the 3 week recovery period. In contrast, retinal degeneration mediated by directly induced apoptosis did not recover in the 3 week recovery period. Dystrophic rod photoreceptors with truncated rod outer segments were identified as the likely source of rod photoreceptor regeneration in the P23H retinas. These dystrophic photoreceptors remain metabolically active despite having lost most of their outer segments.


Amino Acid Substitution , Mutation , Retinal Degeneration/genetics , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/genetics , Rhodopsin/metabolism , Amino Acid Substitution/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Histidine/genetics , Mutation/genetics , Nerve Regeneration/genetics , Proline/genetics , Retinal Degeneration/metabolism , Retinal Rod Photoreceptor Cells/physiology , Retinitis Pigmentosa/metabolism , Rhodopsin/genetics , Rhodopsin/physiology , Xenopus laevis
14.
Vision Res ; 51(21-22): 2304-11, 2011 Nov.
Article En | MEDLINE | ID: mdl-21945483

Photoreceptor guanylate cyclase (GC1) is a transmembrane protein and responsible for synthesis of cGMP, the secondary messenger of phototransduction. It consists of an extracellular domain, a single transmembrane domain, and an intracellular domain. It is unknown how GC1 targets to the outer segments where it resides. To identify a putative GC1 targeting signal, we generated a series of peripheral membrane and transmembrane constructs encoding extracellular and intracellular mouse GC1 fragments fused to EGFP. The constructs were expressed in Xenopus laevis rod photoreceptors under the control of the rhodopsin promoter. We examined the localization of GFP-GC1 fusion proteins containing the complete GC1 sequence, or partial GC1 sequences, which were membrane-associated via either the GC1 transmembrane domain or the rhodopsin C-terminal palmitoyl chains. Full-length GFP-GC1 targeted to the rod outer segment disk rims. As a group, fusion proteins containing the entire cytoplasmic domain of GC1 targeted to the OS, whereas other fusion proteins containing portions of the cytoplasmic or the extracellular domains did not. We conclude that GC1 likely has no single linear peptide-based OS targeting signal. Our results suggest targeting is due to either multiple weak signals in the cytoplasmic domain of GC1, or co-transport to the OS with an accessory protein.


Guanylate Cyclase/metabolism , Receptors, Cell Surface/metabolism , Rod Cell Outer Segment/metabolism , Xenopus laevis/metabolism , Animals , Animals, Genetically Modified , Cell Membrane/metabolism , Membrane Fusion Proteins/metabolism , Molecular Sequence Data , Protein Sorting Signals/genetics , Protein Sorting Signals/physiology , Xenopus laevis/genetics
15.
Adv Exp Med Biol ; 664: 509-15, 2010.
Article En | MEDLINE | ID: mdl-20238053

We have recently developed transgenic X. laevis models of retinitis pigmentosa based on the rhodopsin P23H mutation in the context of rhodopsin cDNAs derived from several different species. The mutant rhodopsin in these animals is expressed at low levels, with levels of export from the endoplasmic reticulum to the outer segment that depend on the cDNA context. Retinal degeneration in these models demonstrates varying degrees of light dependence, with the highest light dependence coinciding with the highest ER export efficiency. Rescue of light dependent retinal degeneration by dark rearing is in turn dependent on the capacity of the mutant rhodopsin to bind chromophore. Our results indicate that rhodopsin chromophore can act in vivo as a pharmacological chaperone for P23H rhodopsin, and that light-dependent retinal degeneration caused by P23H rhodopsin is due to reduced chromophore binding.


Disease Models, Animal , Light , Retinal Degeneration/complications , Retinitis Pigmentosa/complications , Xenopus laevis , Animals , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/radiation effects , Models, Biological , Mutant Proteins/metabolism , Retinal Degeneration/pathology , Retinitis Pigmentosa/pathology , Rhodopsin/genetics , Rhodopsin/metabolism
16.
Invest Ophthalmol Vis Sci ; 51(3): 1327-34, 2010 Mar.
Article En | MEDLINE | ID: mdl-19933196

PURPOSE: To characterize the influence of light and vitamin A on retinal degeneration in an animal model of retinitis pigmentosa caused by the rhodopsin P23H mutation. METHODS: Retinal degeneration was examined in transgenic Xenopus laevis expressing P23H rhodopsin, in which retinal degeneration is completely rescued by preventing light exposure. The sensitivity of this retinal degeneration to varying intensities, wavelengths, and durations of light exposure, and to vitamin A deprivation was characterized. RESULTS: Green light was the most effective inducer of retinal degeneration in this model. Retinal degeneration was induced by prolonged exposure to green light and was prevented by filters that block short wavelengths. Reducing the duration of light exposure prevented retinal degeneration, even when the light intensity was proportionally increased. Vitamin A deprivation also induced retinal degeneration associated with defects in P23H rhodopsin biosynthesis. Vitamin A deprivation did not cause retinal degeneration in nontransgenic animals. CONCLUSIONS: The mechanism of retinal degeneration in this animal model of RP involves the interaction of light with rhodopsin rather than with free chromophore or bleached rhodopsin. These results may explain the clinical benefits of vitamin A for patients with retinitis pigmentosa and may indicate that pharmacological chaperones are a viable approach to RP therapy. Results also suggest strategies for minimizing RD in patients through controlling light exposure duration or wavelengths.


Light , Mutation, Missense , Radiation Injuries, Experimental/etiology , Retinal Degeneration/etiology , Rhodopsin/genetics , Rhodopsin/radiation effects , Vitamin A Deficiency/complications , Animals , Animals, Genetically Modified , Male , Microscopy, Confocal , Photometry , Radiation Injuries, Experimental/genetics , Retina/radiation effects , Retinal Degeneration/genetics , Transgenes , Vitamin A/physiology , Xenopus laevis
17.
J Neurosci ; 29(48): 15145-54, 2009 Dec 02.
Article En | MEDLINE | ID: mdl-19955366

Several mutations in the N terminus of the G-protein-coupled receptor rhodopsin disrupt NXS/T consensus sequences for N-linked glycosylation (located at N2 and N15) and cause sector retinitis pigmentosa in which the inferior retina preferentially degenerates. Here we examined the role of rhodopsin glycosylation in biosynthesis, trafficking, and retinal degeneration (RD) using transgenic Xenopus laevis expressing glycosylation-defective human rhodopsin mutants. Although mutations T4K and T4N caused RD, N2S and T4V did not, demonstrating that glycosylation at N2 was not required for photoreceptor viability. In contrast, similar mutations eliminating glycosylation at N15 (N15S and T17M) caused rod death. Expression of T17M was more toxic than T4K to transgenic photoreceptors, further suggesting that glycosylation at N15 plays a more important physiological role than glycosylation at N2. Together, these results indicate that the structure of the rhodopsin N terminus must be maintained by an appropriate amino acid sequence surrounding N2 and may require a carbohydrate moiety at N15. The mutant rhodopsins were rendered less toxic in their dark inactive states, because RD was abolished or significantly reduced when transgenic tadpoles expressing T4K, T17M, and N2S/N15S were protected from light exposure. Regardless of their effect on rod viability, all of the mutants primarily localized to the outer segment and Golgi and showed little or no endoplasmic reticulum accumulation. Thus, glycosylation was not crucial for rhodopsin biosynthesis or trafficking. Interestingly, expression of similar bovine rhodopsin mutants did not cause rod cell death, possibly attributable to greater stability of bovine rhodopsin.


Light/adverse effects , Protein Folding , Retinal Degeneration/metabolism , Rhodopsin/metabolism , Amino Acids/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Glycosylation , Green Fluorescent Proteins/genetics , Humans , Larva , Mutation/genetics , Photoreceptor Cells, Vertebrate/metabolism , Protein Transport/genetics , Protein Transport/physiology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Rhodopsin/genetics , Xenopus
18.
EMBO J ; 28(3): 183-92, 2009 Feb 04.
Article En | MEDLINE | ID: mdl-19153612

Dysfunctions of primary cilia and cilia-derived sensory organelles underlie a multitude of human disorders, including retinal degeneration, yet membrane targeting to the cilium remains poorly understood. Here, we show that the newly identified ciliary targeting VxPx motif present in rhodopsin binds the small GTPase Arf4 and regulates its association with the trans-Golgi network (TGN), which is the site of assembly and function of a ciliary targeting complex. This complex is comprised of two small GTPases, Arf4 and Rab11, the Rab11/Arf effector FIP3, and the Arf GTPase-activating protein ASAP1. ASAP1 mediates GTP hydrolysis on Arf4 and functions as an Arf4 effector that regulates budding of post-TGN carriers, along with FIP3 and Rab11. The Arf4 mutant I46D, impaired in ASAP1-mediated GTP hydrolysis, causes aberrant rhodopsin trafficking and cytoskeletal and morphological defects resulting in retinal degeneration in transgenic animals. As the VxPx motif is present in other ciliary membrane proteins, the Arf4-based targeting complex is most likely a part of conserved machinery involved in the selection and packaging of the cargo destined for delivery to the cilium.


ADP-Ribosylation Factors/metabolism , Cilia/metabolism , Rhodopsin/chemistry , Rhodopsin/metabolism , Xenopus Proteins/metabolism , Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Animals, Genetically Modified , Cilia/ultrastructure , GTPase-Activating Proteins/metabolism , Guanosine Triphosphate/metabolism , Hydrolysis , I-kappa B Kinase/chemistry , I-kappa B Kinase/metabolism , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Molecular Sequence Data , Mutant Proteins/metabolism , Organ Specificity , Protein Binding , Protein Sorting Signals , Protein Structure, Tertiary , Protein Transport , Retinal Degeneration/metabolism , Xenopus/genetics , rab GTP-Binding Proteins/metabolism , trans-Golgi Network/ultrastructure
19.
Invest Ophthalmol Vis Sci ; 50(2): 885-92, 2009 Feb.
Article En | MEDLINE | ID: mdl-18836175

PURPOSE: Because of their high cone/rod ratio, Xenopus laevis may be a useful system for examining rod-cone interactions during retinal degeneration and mechanisms that underlie secondary cone degeneration. The authors developed an inducible model of retinitis pigmentosa (RP) in X. laevis to investigate these issues. METHODS: The authors generated transgenic X. laevis that express a modified caspase-9 (iCasp9) under the control of the X. laevis rod opsin promoter. iCasp9 is activated by the compound AP20187, resulting in an apoptotic cascade. Confocal microscopy, Western blot analysis, and electroretinography (ERG) were used to determine the effects of AP20187 on transgenic retinas. RESULTS: AP20187 induced rod cell apoptosis in transgenic tadpoles and postmetamorphic frogs. Longitudinal results indicate rod cell death led to cone cell dysfunction within 3 months; however, cone function was reinstated after 6 months. Returning cone function may be associated with increased numbers of morphologically normal cone cells and thickening of the inner nuclear layer. CONCLUSIONS: These studies indicate that X. laevis may be a useful system for examining cone dysfunction associated with rod death in RP and longer term regeneration of cone responses. This inducible model of RP is unique in that rod death proceeds through a well-understood mechanism, rod death can be carefully controlled to occur at any stage of development, and the stimulus for rod death can be removed at any time.


Apoptosis/drug effects , Retina/physiopathology , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/pathology , Tacrolimus/analogs & derivatives , Animals , Animals, Genetically Modified , Blotting, Western , Caspase 9/genetics , Caspase 9/metabolism , Disease Models, Animal , Electroretinography , Enzyme Activation , Gene Expression Regulation, Enzymologic/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Microscopy, Confocal , Opsins/genetics , Promoter Regions, Genetic , Retina/enzymology , Retinal Cone Photoreceptor Cells/enzymology , Retinal Rod Photoreceptor Cells/enzymology , Retinitis Pigmentosa/enzymology , Retinitis Pigmentosa/genetics , Tacrolimus/pharmacology , Xenopus laevis/genetics
20.
Nucleic Acids Res ; 36(20): 6523-34, 2008 Nov.
Article En | MEDLINE | ID: mdl-18927113

X-linked juvenile retinoschisis is a heritable condition of the retina in males caused by mutations in the RS1 gene. Still, the cellular function and retina-specific expression of RS1 are poorly understood. To address the latter issue, we characterized the minimal promoter driving expression of RS1 in the retina. Binding site prediction, site-directed mutagenesis, and reporter assays suggest an essential role of two nearby cone-rod homeobox (CRX)-responsive elements (CRE) in the proximal -177/+32 RS1 promoter. Chromatin immunoprecipitation associates the RS1 promoter in vivo with CRX, the coactivators CBP, P300, GCN5 and acetylated histone H3. Transgenic Xenopus laevis expressing a green fluorescent protein (GFP) reporter under the control of RS1 promoter sequences show that the -177/+32 fragment drives GFP expression in photoreceptors and bipolar cells. Mutating either of the two conserved CRX binding sites results in strongly decreased RS1 expression. Despite the presence of sequence motifs in the promoter, NRL and NR2E3 appear not to be essential for RS1 expression. Together, our in vitro and in vivo results indicate that two CRE sites in the minimal RS1 promoter region control retinal RS1 expression and establish CRX as a key factor driving this expression.


Eye Proteins/genetics , Homeodomain Proteins/metabolism , Promoter Regions, Genetic , Retina/metabolism , Trans-Activators/metabolism , Acetylation , Animals , Base Sequence , Binding Sites , Cell Line , Dogs , Eye Proteins/biosynthesis , Histones/metabolism , Humans , Mice , Molecular Sequence Data , Photoreceptor Cells, Vertebrate/metabolism , Response Elements , Retinal Bipolar Cells/metabolism , Transcriptional Activation , Xenopus laevis , p300-CBP Transcription Factors/metabolism
...