Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Transl Vis Sci Technol ; 11(5): 24, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35604672

ABSTRACT

Purpose: Optogenetic gene therapy to render remaining retinal cells light-sensitive in end-stage retinal degeneration is a promising strategy for treatment of individuals blind because of a variety of different inherited retinal degenerations. The clinical trials currently in progress focus on delivery of optogenetic genes to ganglion cells. Delivery of optogenetic molecules to cells in the outer neural retina is predicted to be even more advantageous because it harnesses more of the retinal circuitry. However, this approach has not yet been tested in large animal models. For this reason, we evaluated the safety and efficacy of optogenetic therapy targeting remaining diseased cone photoreceptors in the Rcd1 dog model of retinitis pigmentosa. Methods: Imaging and measures of retinal function and functional vision were carried out, as well as terminal studies evaluating multi-electrode array recordings and histology. Results: Animals remained healthy and active throughout the study and showed improved retinal and visual function as assessed by electroretinography and visual-evoked potentials, improved navigational vision, and improved function of cone photoreceptors and the downstream retinal circuitry. Conclusions: The findings demonstrate that an optogenetic approach targeting the outer retina in a blind large animal model can partially restore vision. Translational Relevance: This work has translational relevance because the approach could potentially be extrapolated to treat humans who are totally blind because of retinal degenerative disease.


Subject(s)
Dependovirus , Retinal Degeneration , Animals , Dependovirus/genetics , Dogs , Optogenetics/methods , Retina , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Degeneration/therapy , Vision, Ocular
2.
Mol Ther ; 26(6): 1581-1593, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29673930

ABSTRACT

Most genetically distinct inherited retinal degenerations are primary photoreceptor degenerations. We selected a severe early onset form of Leber congenital amaurosis (LCA), caused by mutations in the gene LCA5, in order to test the efficacy of gene augmentation therapy for a ciliopathy. The LCA5-encoded protein, Lebercilin, is essential for the trafficking of proteins and vesicles to the photoreceptor outer segment. Using the AAV serotype AAV7m8 to deliver a human LCA5 cDNA into an Lca5 null mouse model of LCA5, we show partial rescue of retinal structure and visual function. Specifically, we observed restoration of rod-and-cone-driven electroretinograms in about 25% of injected eyes, restoration of pupillary light responses in the majority of treated eyes, an ∼20-fold decrease in target luminance necessary for visually guided behavior, and improved retinal architecture following gene transfer. Using LCA5 patient-derived iPSC-RPEs, we show that delivery of the LCA5 cDNA restores lebercilin protein and rescues cilia quantity. The results presented in this study support a path forward aiming to develop safety and efficacy trials for gene augmentation therapy in human subjects with LCA5 mutations. They also provide the framework for measuring the effects of intervention in ciliopathies and other severe, early-onset blinding conditions.


Subject(s)
Blindness/metabolism , Blindness/therapy , Dependovirus/genetics , Genetic Therapy/methods , Animals , Electroretinography , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Humans , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/therapy , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism
3.
PLoS One ; 11(11): e0166348, 2016.
Article in English | MEDLINE | ID: mdl-27893831

ABSTRACT

Complement dysregulation plays a key role in the pathogenesis of age-related macular degeneration (AMD), but the specific mechanisms are incompletely understood. Complement also potentiates retinal degeneration in the murine light damage model. To test the retinal function of CD59a, a complement inhibitor, CD59a knockout (KO) mice were used for light damage (LD) experiments. Retinal degeneration and function were compared in WT versus KO mice following light damage. Gene expression changes, endoplasmic reticulum (ER) stress, and glial cell activation were also compared. At baseline, the ERG responses and rhodopsin levels were lower in CD59aKO compared to wild-type (WT) mice. Following LD, the ERG responses were better preserved in CD59aKO compared to WT mice. Correspondingly, the number of photoreceptors was higher in CD59aKO retinas than WT controls after LD. Under normal light conditions, CD59aKO mice had higher levels than WT for GFAP immunostaining in Müller cells, mRNA and protein levels of two ER-stress markers, and neurotrophic factors. The reduction in photon capture, together with the neurotrophic factor upregulation, may explain the structural and functional protection against LD in the CD59aKO.


Subject(s)
CD59 Antigens/genetics , Light , Photoreceptor Cells, Vertebrate/radiation effects , Retinal Degeneration/pathology , Animals , CD59 Antigens/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Electroretinography , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/radiation effects , Ependymoglial Cells/metabolism , Eye Enucleation , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neuroglia/radiation effects , Phagocytosis/radiation effects , Photoreceptor Cells, Vertebrate/metabolism , RNA, Messenger/metabolism , Retina/diagnostic imaging , Retina/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/veterinary , Retinaldehyde/analysis , Rhodopsin/genetics , Rhodopsin/metabolism , Up-Regulation/radiation effects
4.
J Physiol ; 593(7): 1531-50, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25416620

ABSTRACT

Heterotrimeric G-proteins (comprising Gα and Gßγ subunits) are critical for coupling of metabotropic receptors to their downstream effectors. In the retina, glutamate released from photoreceptors in the dark activates metabotropic glutamate receptor 6 (mGluR6) receptors in ON bipolar cells; this leads to activation of Go , closure of transient receptor potential melastatin 1 channels and hyperpolarization of these cells. Go comprises Gαo , Gß3 and a Gγ. The best Gγ candidate is Gγ13, although functional data to support this are lacking. Thus, we tested Gγ13 function by generating Gng13(-/-) knockout (KO) mice, recording electroretinograms (ERG) and performing immunocytochemical staining. The amplitude of scotopic ERG b-waves in KO mice was lower than in wild-type (WT) mice. Furthermore, in both KO and WT mice, the ERG b-wave decreased with age; this decrease was much more pronounced in KO mice. By contrast, the photopic ERG b-waves in KO mice were hardly affected at any age. In KO mice retinas, immunostaining for Gß3 and for the GTPase activating proteins RGS7, RGS11, R9AP and Gß5 decreased significantly in rod bipolar cells but not in ON cone bipolar cells. Staining for Gαo and certain other cascade elements decreased only slightly. Analysis of our ON bipolar cDNA library showed that these cells express mRNAs for Gγ5, Gγ10 and Gγ11. Quantitative RT-PCR of retinal cDNA showed greater values for these transcripts in retinas of KO mice, although the difference was not significant. Our results suggest that Gγ13 contributes to mGluR6 signalling in rod bipolar cells more than in ON cone bipolar cells, and that this contribution includes both coupling the receptor and maintaining a stable localization of the mGluR6-related cascade elements.


Subject(s)
Heterotrimeric GTP-Binding Proteins/physiology , Receptors, Metabotropic Glutamate/physiology , Retinal Bipolar Cells/physiology , Animals , Electroretinography , Female , Heterotrimeric GTP-Binding Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout
5.
Front Mol Neurosci ; 7: 60, 2014.
Article in English | MEDLINE | ID: mdl-25018695

ABSTRACT

The ubiquitous second messenger cGMP is synthesized by guanylyl cyclase and hydrolyzed by phosphodiesterase (PDE). cGMP mediates numerous signaling pathways in multiple tissues. In the retina, cGMP regulates signaling in nearly every cell class including photoreceptors, bipolar cells, amacrine cells, and ganglion cells. In order to understand the specific role of cGMP and its regulating enzymes in different cell types, it is first necessary to localize these components and dissect their influence on the circuits. Here we tested the contribution of PDE9A to retinal processing by recording the electroretinograms (ERG) of PDE9A (™/™) (KO) mice and by localizing the enzyme. We found that while the scotopic ERG of KO was the same as that of wild type (WT) in both amplitude and kinetics, the photopic ERG was greatly affected. The greatest effect was on the recovery of the b-wave; the falling phase and the b-wave duration were significantly longer in the KO mice for all photopic stimuli (UV, green, or saturating white flashes). The rising phase was slower in KO than in WT for UV and green stimuli. For certain stimuli, amplitudes of both the a- and b-waves were smaller than in WT. Using Lac-Z expression in KO retinas as a reporter for PDE9A expression pattern, we found that PDE9A is localized to GABA-positive and GABA-negative amacrine cells, and likely also to certain types of ganglion cells. Our results indicate that PDE9A, by controlling the level of cGMP, modulates inhibitory processes within the cone pathway. We speculate that these circuits involve NO/cGMP signaling pathways.

6.
J Neurosci ; 33(12): 5182-94, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23516284

ABSTRACT

Mammalian cones respond to light by closing a cGMP-gated channel via a cascade that includes a heterotrimeric G-protein, cone transducin, comprising Gαt2, Gß3 and Gγt2 subunits. The function of Gßγ in this cascade has not been examined. Here, we investigate the role of Gß3 by assessing cone structure and function in Gß3-null mouse (Gnb3(-/-)). We found that Gß3 is required for the normal expression of its partners, because in the Gnb3(-/-) cone outer segments, the levels of Gαt2 and Gγt2 are reduced by fourfold to sixfold, whereas other components of the cascade remain unaltered. Surprisingly, Gnb3(-/-) cones produce stable responses with normal kinetics and saturating response amplitudes similar to that of the wild-type, suggesting that cone phototransduction can function efficiently without a Gß subunit. However, light sensitivity was reduced by approximately fourfold in the knock-out cones. Because the reduction in sensitivity was similar in magnitude to the reduction in Gαt2 level in the cone outer segment, we conclude that activation of Gαt2 in Gnb3(-/-) cones proceeds at a rate approximately proportional to its outer segment concentration, and that activation of phosphodiesterase and downstream cascade components is normal. These results suggest that the main role of Gß3 in cones is to establish optimal levels of transducin heteromer in the outer segment, thereby indirectly contributing to robust response properties.


Subject(s)
Heterotrimeric GTP-Binding Proteins/genetics , Retinal Cone Photoreceptor Cells/physiology , Transducin/genetics , Vision, Ocular/physiology , Animals , Color , Female , GABA Plasma Membrane Transport Proteins/genetics , Green Fluorescent Proteins/genetics , Heterotrimeric GTP-Binding Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Photic Stimulation , Retinal Photoreceptor Cell Outer Segment/physiology , Transducin/physiology , Ultraviolet Rays
7.
J Neurosci ; 32(33): 11343-55, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22895717

ABSTRACT

Heterotrimeric G-proteins, comprising Gα and Gßγ subunits, couple metabotropic receptors to various downstream effectors and contribute to assembling and trafficking receptor-based signaling complexes. A G-protein ß subunit, Gß(3), plays a critical role in several physiological processes, as a polymorphism in its gene is associated with a risk factor for several disorders. Retinal ON bipolar cells express Gß(3), and they provide an excellent system to study its role. In the ON bipolar cells, mGluR6 inverts the photoreceptor's signal via a cascade in which glutamate released from photoreceptors closes the TRPM1 channel. This cascade is essential for vision since deficiencies in its proteins lead to complete congenital stationary night blindness. Here we report that Gß(3) participates in the G-protein heterotrimer that couples mGluR6 to TRPM1. Gß(3) deletion in mouse greatly reduces the light response under both scotopic and photopic conditions, but it does not eliminate it. In addition, Gß(3) deletion causes mislocalization and downregulation of most cascade elements and modulators. Furthermore, Gß(3) may play a role in synaptic maintenance since in its absence, the number of invaginating rod bipolar dendrites is greatly reduced, a deficit that was not observed at 3 weeks, the end of the developmental period.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , Gene Expression Regulation/genetics , Retinal Bipolar Cells/metabolism , Synapses/physiology , Animals , Choline O-Acetyltransferase/metabolism , Dendrites/ultrastructure , Electric Stimulation , Electroretinography , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein beta Subunits/deficiency , GTPase-Activating Proteins/metabolism , Green Fluorescent Proteins/genetics , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Immunoprecipitation , In Vitro Techniques , Light , Membrane Potentials/drug effects , Membrane Potentials/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Photic Stimulation , Propionates/pharmacology , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/deficiency , Receptors, Metabotropic Glutamate/genetics , Retina/cytology , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/ultrastructure , Retinal Cone Photoreceptor Cells/metabolism , Synapses/genetics , Synapses/metabolism , Synapses/ultrastructure , TRPM Cation Channels/metabolism , Visual Pathways/physiology
9.
Invest Ophthalmol Vis Sci ; 52(2): 959-68, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21051716

ABSTRACT

PURPOSE: Iron-induced oxidative stress may exacerbate age-related macular degeneration (AMD). Ceruloplasmin/Hephaestin double-knockout (DKO) mice with age-dependent retinal iron accumulation and some features of AMD were used to test retinal protection by the oral iron chelator deferiprone (DFP). METHODS: Cultured retinal pigment epithelial (ARPE-19) cells and mice were treated with DFP. Transferrin receptor mRNA (Tfrc), an indicator of iron levels, was quantified by qPCR. In mice, retinal oxidative stress was assessed by mass spectrometry, and degeneration by histology and electroretinography. RESULTS: DFP at 60 µM decreased labile iron in ARPE-19 cells, increasing Tfrc and protecting 70% of cells against a lethal dose of H(2)O(2). DFP 1 mg/mL in drinking water increased retinal Tfrc mRNA 2.7-fold after 11 days and also increased transferrin receptor protein. In DKOs, DFP over 8 months decreased retinal iron levels to 72% of untreated mice, diminished retinal oxidative stress to 70% of the untreated level, and markedly ameliorated retinal degeneration. DFP was not retina toxic in wild-type (WT) or DKO mice, as assessed by histology and electroretinography. CONCLUSIONS: Oral DFP was not toxic to the mouse retina. It diminished retinal iron levels and oxidative stress and protected DKO mice against iron overload-induced retinal degeneration. Further testing of DFP for retinal disease involving oxidative stress is warranted.


Subject(s)
Iron Chelating Agents/administration & dosage , Iron Overload/prevention & control , Pyridones/administration & dosage , Retinal Degeneration/prevention & control , Retinal Pigment Epithelium/drug effects , Administration, Oral , Animals , Antigens, CD/metabolism , Cell Death , Cell Line , Ceruloplasmin/genetics , Deferiprone , Electroretinography , Female , Fluorescent Antibody Technique, Indirect , Humans , Hydrogen Peroxide/toxicity , Iron Chelating Agents/pharmacology , Iron Overload/metabolism , Iron Overload/pathology , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Oxidative Stress/drug effects , Pyridones/pharmacology , RNA, Messenger/genetics , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Pigment Epithelium/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
J Neurosci ; 30(9): 3347-57, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-20203194

ABSTRACT

Maturation of the mammalian nervous system requires adequate provision of thyroid hormone and mechanisms that enhance tissue responses to the hormone. Here, we report that the development of cones, the photoreceptors for daylight and color vision, requires protection from thyroid hormone by type 3 deiodinase, a thyroid hormone-inactivating enzyme. Type 3 deiodinase, encoded by Dio3, is expressed in the immature mouse retina. In Dio3(-/-) mice, approximately 80% of cones are lost through neonatal cell death. Cones that express opsin photopigments for response to both short (S) and medium-long (M) wavelength light are lost. Rod photoreceptors, which mediate dim light vision, remain essentially intact. Excessive thyroid hormone in wild-type pups also eliminates cones. Cone loss is mediated by cone-specific thyroid hormone receptor beta2 (TRbeta2) as deletion of TRbeta2 rescues cones in Dio3(-/-) mice. However, rescued cones respond to short but not longer wavelength light because TRbeta2 under moderate hormonal stimulation normally induces M opsin and controls the patterning of M and S opsins over the retina. The results suggest that type 3 deiodinase limits hormonal exposure of the cone to levels that safeguard both cone survival and the patterning of opsins that is required for cone function.


Subject(s)
Iodide Peroxidase/genetics , Retina/enzymology , Retina/growth & development , Retinal Cone Photoreceptor Cells/enzymology , Thyroid Hormones/metabolism , Animals , Cell Death/genetics , Cell Differentiation/genetics , Cell Survival/genetics , Female , Gene Expression Regulation, Developmental/genetics , Light , Male , Mice , Mice, Knockout , Opsins/metabolism , Photic Stimulation , Retina/cytology , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/radiation effects , Thyroid Hormone Receptors beta/metabolism , Vision, Ocular/genetics
11.
Lancet ; 374(9701): 1597-605, 2009 Nov 07.
Article in English | MEDLINE | ID: mdl-19854499

ABSTRACT

BACKGROUND: Gene therapy has the potential to reverse disease or prevent further deterioration of vision in patients with incurable inherited retinal degeneration. We therefore did a phase 1 trial to assess the effect of gene therapy on retinal and visual function in children and adults with Leber's congenital amaurosis. METHODS: We assessed the retinal and visual function in 12 patients (aged 8-44 years) with RPE65-associated Leber's congenital amaurosis given one subretinal injection of adeno-associated virus (AAV) containing a gene encoding a protein needed for the isomerohydrolase activity of the retinal pigment epithelium (AAV2-hRPE65v2) in the worst eye at low (1.5 x 10(10) vector genomes), medium (4.8 x 10(10) vector genomes), or high dose (1.5 x 10(11) vector genomes) for up to 2 years. FINDINGS: AAV2-hRPE65v2 was well tolerated and all patients showed sustained improvement in subjective and objective measurements of vision (ie, dark adaptometry, pupillometry, electroretinography, nystagmus, and ambulatory behaviour). Patients had at least a 2 log unit increase in pupillary light responses, and an 8-year-old child had nearly the same level of light sensitivity as that in age-matched normal-sighted individuals. The greatest improvement was noted in children, all of whom gained ambulatory vision. The study is registered with ClinicalTrials.gov, number NCT00516477. INTERPRETATION: The safety, extent, and stability of improvement in vision in all patients support the use of AAV-mediated gene therapy for treatment of inherited retinal diseases, with early intervention resulting in the best potential gain. FUNDING: Center for Cellular and Molecular Therapeutics at the Children's Hospital of Philadelphia, Foundation Fighting Blindness, Telethon, Research to Prevent Blindness, F M Kirby Foundation, Mackall Foundation Trust, Regione Campania Convenzione, European Union, Associazione Italiana Amaurosi Congenita di Leber, Fund for Scientific Research, Fund for Research in Ophthalmology, and National Center for Research Resources.


Subject(s)
Carrier Proteins/genetics , Eye Proteins/genetics , Genetic Therapy/methods , Optic Atrophy, Hereditary, Leber/therapy , Adolescent , Adult , Age Factors , Blindness/congenital , Blindness/genetics , Child , Dark Adaptation , Dependovirus/genetics , Disease Progression , Dose-Response Relationship, Drug , Electroretinography , Female , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Humans , Injections , Male , Mutation/genetics , Nystagmus, Physiologic , Optic Atrophy, Hereditary, Leber/diagnosis , Optic Atrophy, Hereditary, Leber/genetics , Safety , Treatment Outcome , Visual Acuity , Young Adult , cis-trans-Isomerases
12.
N Engl J Med ; 358(21): 2240-8, 2008 May 22.
Article in English | MEDLINE | ID: mdl-18441370

ABSTRACT

Leber's congenital amaurosis (LCA) is a group of inherited blinding diseases with onset during childhood. One form of the disease, LCA2, is caused by mutations in the retinal pigment epithelium-specific 65-kDa protein gene (RPE65). We investigated the safety of subretinal delivery of a recombinant adeno-associated virus (AAV) carrying RPE65 complementary DNA (cDNA) (ClinicalTrials.gov number, NCT00516477 [ClinicalTrials.gov]). Three patients with LCA2 had an acceptable local and systemic adverse-event profile after delivery of AAV2.hRPE65v2. Each patient had a modest improvement in measures of retinal function on subjective tests of visual acuity. In one patient, an asymptomatic macular hole developed, and although the occurrence was considered to be an adverse event, the patient had some return of retinal function. Although the follow-up was very short and normal vision was not achieved, this study provides the basis for further gene therapy studies in patients with LCA.


Subject(s)
Blindness/therapy , Carrier Proteins/genetics , Eye Proteins/genetics , Genetic Therapy , Genetic Vectors , Retinal Degeneration/therapy , Adult , Blindness/congenital , Blindness/genetics , Blindness/pathology , DNA, Complementary , Dependovirus/genetics , Gene Transfer Techniques , Humans , Injections , Mutation , Promoter Regions, Genetic , Reflex, Pupillary , Retina/pathology , Retinal Degeneration/congenital , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Visual Acuity , cis-trans-Isomerases
13.
Invest Ophthalmol Vis Sci ; 49(3): 1126-35, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18326740

ABSTRACT

PURPOSE: To define rod and cone function further in terms of visual cycle mechanism, the retinal phenotype resulting from Rpe65 (retinoid isomerase I) deficiency in Nrl(-)(/)(-) mice having a single class of photoreceptors resembling wild-type cones was characterized and outcomes of retinoid supplementation evaluated. METHODS: Rpe65(-)(/)(-)/Nrl(-)(/)(-) mice were generated by breeding Rpe65(-)(/)(-) and Nrl(-)(/)(-) strains. Retinal histology, protein expression, retinoid content, and electroretinographic (ERG) responses were evaluated before and after treatment with 11-cis retinal by intraperitoneal injection. Results Retinas of young Rpe65(-)(/-)/Nrl(-)(/-) mice exhibited normal lamination, but lacked intact photoreceptor outer segments at all ages examined. Rpe65, Nrl, and rhodopsin were not detected, and S-opsin and M/L-opsin levels were reduced. Retinyl esters were the only retinoids present. In contrast, Nrl(-)(/)(-) mice exhibited decreased levels of retinaldehydes and retinyl esters, and elevated levels of retinols. ERG responses were elicited from Rpe65(-)(/-)/Nrl(-)(/-) mice only at the two highest intensities over a 4-log-unit range. Significant retinal thinning and outer nuclear layer loss occurred in Rpe65(-)(/-)/Nrl(-)(/-) mice with aging. Administration of exogenous 11-cis retinal did not rescue retinal morphology or markedly improve ERG responses. CONCLUSIONS: The findings provide clarification of reported cone loss of function in Rpe65(-)(/-)/Nrl(-)(/-) mice, now showing that chromophore absence results in destabilized cone outer segments and rapid retinal degeneration. The data support the view that rod-dominant retinas do not have a cone-specific mechanism for 11-cis retinal synthesis and have potential significance for therapeutic strategies for rescue of cone-rich retinal regions affected by disease in the aging human population.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , Carrier Proteins/physiology , Eye Proteins/physiology , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Degeneration/metabolism , Retinaldehyde/biosynthesis , Animals , Blotting, Western , Chromatography, High Pressure Liquid , Dark Adaptation , Electroretinography , Female , Fluorescent Antibody Technique, Indirect , Genotype , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/physiopathology , Retinal Degeneration/drug therapy , Retinal Degeneration/physiopathology , Retinaldehyde/administration & dosage , Retinoids/metabolism , Rod Opsins/metabolism , cis-trans-Isomerases
14.
Biochemistry ; 44(29): 9880-8, 2005 Jul 26.
Article in English | MEDLINE | ID: mdl-16026160

ABSTRACT

RPE65, a protein expressed in cells of the retinal pigment epithelium of the eye, is essential for the synthesis by isomerohydrolase of 11-cis-retinal, the chromophore of rod and cone opsins. Recent work has established that RPE65 is a retinyl ester binding protein, and as all-trans-retinyl esters are the substrate for isomerohydrolase activity, the hypothesis has emerged that RPE65 serves to deliver substrate to this enzyme or complex. We bred mice with five distinct combinations of the RPE65 Leu450/Met450 variants (Leu/Leu, Met/Met, Leu/Met, Leu/-, and Met/-), measured in mice of each genotype the mole quantity of RPE65 per eye, and measured the initial rate of rhodopsin regeneration after a nearly complete bleach of rhodopsin to estimate the maximum rate of 11-cis-retinal synthesis in vivo. The quantity of RPE65 per eye ranged from 5.7 pmol (Balb/c) to 0.32 pmol (C57BL/6N x Rpe65(-)(/)(-)); the initial rate of rhodopsin regeneration was a Michaelis function of RPE65, where V(max) = 18 pmol/min per eye and K(m) = 1.7 pmol, and not dependent on the Leu450/Met450 variant. At RPE65 levels well below the K(m), the rate of production of 11-cis-retinal per RPE65 molecule was approximately 10 min(-)(1). Thus, the results imply that as a chaperone each RPE65 molecule can deliver retinyl ester to the isomerohydrolase at a rate of 10 molecules/min; should RPE65 itself be identified as the isomerase, each copy must be able to produce at least 10 molecules of 11-cis-retinal per minute.


Subject(s)
Eye Proteins/biosynthesis , Eye Proteins/chemistry , Molecular Chaperones/biosynthesis , Molecular Chaperones/chemistry , Pigment Epithelium of Eye/chemistry , Pigment Epithelium of Eye/metabolism , Retinaldehyde/biosynthesis , Animals , Carrier Proteins , Crosses, Genetic , Esters , Eye Proteins/genetics , Immunoblotting , Kinetics , Leucine/genetics , Methionine/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , Pigment Epithelium of Eye/cytology , RNA, Messenger/biosynthesis , Rhodopsin/biosynthesis , Substrate Specificity , cis-trans-Isomerases/chemistry , cis-trans-Isomerases/metabolism
15.
Invest Ophthalmol Vis Sci ; 46(6): 2156-67, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15914637

ABSTRACT

PURPOSE: To test the hypothesis that Nrl(-)(/)(-) photoreceptors are cones, by comparing them with WT rods and cones using morphological, molecular, histochemical, and electrophysiological criteria. METHODS: The photoreceptor layer of fixed retinal tissue of 4- to 6-week-old mice was examined in plastic sections by electron microscopy, and by confocal microscopy in frozen sections immunolabeled for the mouse UV-cone pigment and colabeled with PNA. Quantitative immunoblot analysis was used to determine the levels of expression of key cone-specific proteins. Single- and paired-flash methods were used to extract the spectral sensitivity, kinetics, and amplification of the a-wave of the ERG. RESULTS: Outer segments of Nrl(-/-) photoreceptors ( approximately 7 mum) are shorter than those of wild-type (WT) rods ( approximately 25 mum) and cones ( approximately 15 mum); but, like WT cones, they have 25 or more basal discs open to the extracellular space, extracellular matrix sheaths stained by PNA, chromatin "clumping" in their nuclei, and mitochondria two times shorter than rods. Nrl(-/-) photoreceptors express the mouse UV cone pigment, cone transducin, and cone arrestin in amounts expected, given the relative size and density of cones in the two retinas. The ERG a-wave was used to assay the properties of the photocurrent response. The sensitivity of the Nrl(-/-) a-wave is at its maximum at 360 nm, with a secondary mode at 510 nm having approximately one-tenth the maximum sensitivity. These wavelengths are the lambda(max) of the two mouse cone pigments. The time to peak of the dim-flash photocurrent response was approximately 50 ms, more than two times faster than that of rods. CONCLUSIONS: Many morphological, molecular, and electrophysiological features of the Nrl(-/-) photoreceptors are cone-like, and strongly distinguish these cells from rods. This retina provides a model for the investigation of cone function and cone-specific genetic disease.


Subject(s)
DNA-Binding Proteins/physiology , Eye Proteins/physiology , Leucine Zippers/physiology , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/physiology , Animals , Arrestin/metabolism , Basic-Leucine Zipper Transcription Factors , Biomarkers/metabolism , Electrophysiology , Electroretinography , Female , Immunoblotting , Mice , Mice, Inbred BALB C , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Pregnancy , Retinal Pigments/metabolism , Rhodopsin/metabolism , Transducin/metabolism , Vision, Ocular/physiology
16.
Mol Vis ; 11: 1236-45, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16402024

ABSTRACT

PURPOSE: The concentration of enhanced green fluorescent protein (EGFP) in individual photoreceptor cells of live mouse retina was quantified and correlated with physiological measurements of cell function. METHODS: EGFP protein levels in the retinas of mice injected subretinally by either one of two serotypes of adeno-associated virus (AAV; AAV2/5.CMV.EGFP; AAV2/2.CMV.EGFP) were quantified with a photon-counting confocal laser scanning microscope and compared with those of transgenic mice whose retinas expressed EGFP under the beta-actin (pbetaAct) or human L/M-cone opsin (pLMCOps) promoter. Single-cell suction pipette recordings of single rods and whole-field electroretinograms (ERGs) were performed to assess retinal cell function. RESULTS: The highest levels of EGFP (680 microM) were in the retinal pigment epithelium (RPE) cells of the AAV-transduced eyes. Living photoreceptors of pbetaAct.EGFP mice contained 270 microM EGFP, while their bipolars had 440 microM. The cones of pLMCOps.EGFP mice expressed 60 microM protein. The amplitudes of the major components of ERGs were within the normal range for all transgenic animals examined, and single cell recordings from living pbetaAct.EGFP rods were indistinguishable from those of controls. CONCLUSIONS: EGFP levels in individual cells of live mouse retinas can be quantified, so that the efficacy of gene transfer methods can be quantified. Concentrations of several hundred microM are not deleterious to normal function of photoreceptors and bipolar cells. This approach can also be used to quantify levels of biologically active EGFP fusion proteins.


Subject(s)
Gene Transfer Techniques , Green Fluorescent Proteins/pharmacokinetics , Green Fluorescent Proteins/poisoning , Retina/drug effects , Retina/metabolism , Animals , Dependovirus/genetics , Electrophysiology , Electroretinography , Embryo, Mammalian/metabolism , Genetic Vectors , Green Fluorescent Proteins/administration & dosage , Injections , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Osmolar Concentration , Photic Stimulation , Photoreceptor Cells, Vertebrate/metabolism , Retina/cytology , Retina/physiology , Retinal Bipolar Cells/metabolism , Tissue Distribution
17.
Vision Res ; 44(28): 3235-51, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15535992

ABSTRACT

To quantify the rate at which light in a ganzfeld produces photoisomerizations in mouse rods in situ, we measured the rate of rhodopsin bleaching in eyes of recently euthanized mice with fully dilated pupils. The amount of rhodopsin declined as a first-order (exponential) function of the duration of the exposure at the luminance of 920 scot cd m(-2): the rate constants of bleaching were 8.3 x 10(-6) and 2.8 x 10(-5) s(-1) (scot cd(-1)m2)(-1) for C57B1/6 and 129P3/J mice, respectively. When the approximately 3-fold difference in effective areas of the pupils of the mice are taken into consideration, the bleaching rates for both strains become essentially the same, 2.6 x 10(-6) fraction rhodopsin (scot Td s)(-1). Assuming 7 x 10(7) rhodopsin molecules per rod, this bleaching rate yields the result that a flash of 1 scot Td s produces 181 photoisomerizations per rod, a value close to that derived from analysis of the collecting area of the rod for axially propagating light. We measured the electroretinograms of mice of the two strains reared under controlled illumination conditions (2 and 100 lux), and compared their properties, using the calibrations to determine the absolute sensitivities of the b-wave and a-waves. The intensity that produces a half-saturating rod b-wave response is 0.3-0.6 photoisomerizations rod(-1), and the amplification constant of the rod a-wave is 5-6 s(-2) photoisomerization(-1), with little dependence on the strain.


Subject(s)
Retinal Rod Photoreceptor Cells/metabolism , Rhodopsin/metabolism , Albinism, Ocular/metabolism , Albinism, Ocular/pathology , Albinism, Ocular/physiopathology , Animals , Electroretinography/methods , Eye/anatomy & histology , Lighting , Mice , Mice, Inbred C57BL , Models, Biological , Photic Stimulation/methods , Retina/anatomy & histology , Retinal Rod Photoreceptor Cells/physiology , Rod Cell Outer Segment/anatomy & histology , Species Specificity , Vision, Ocular/physiology
18.
Mol Ther ; 10(5): 855-61, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15509503

ABSTRACT

Molecules with neurotrophic activity are being evaluated for treatment of retinitis pigmentosa in animal models. In particular, great interest has been focused recently on erythropoietin (Epo). Evidence of its neurotrophic activity comes mainly from data demonstrating photoreceptor protection in a rodent light-damage model through systemic administration of a recombinant form of this hormone. Our goal was to test whether Epo retinal gene transfer can rescue or delay photoreceptor cell death. We delivered adeno-associated viral vectors encoding Epo intraocularly and, for comparison, intramuscularly to one light-induced and two genetic models of retinal degeneration. Intraocular Epo gene transfer resulted in sustained hormone expression in the eye, which was undetectable systemically. In contrast, Epo intramuscular gene transfer resulted in hormone secretion in the circulation, which was not detected in ocular fluids. The protein secreted from muscle and retina is of the same molecular weight as a commercial recombinant human Epo. Interestingly, following systemic but not intraocular Epo delivery, morphological photoreceptor protection was observed in the light-damage and rds/peripherin (Prph2) models of retinal degeneration. In the light-damage model, the morphological rescue was accompanied by a significant electrophysiological improvement of photoreceptor function. In contrast, no photoreceptor rescue was observed following Epo gene transfer in the rd10 model. This suggests that different apoptotic mechanisms, with varying sensitivities to Epo, occur in different retinal degeneration models. In conclusion, our data support Epo as a neuroprotective agent in some, but not all, retinal degenerations. Further, rescue is observed in specific models after systemic but not intraocular Epo gene transfer.


Subject(s)
Dependovirus/genetics , Erythropoietin/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Retinal Degeneration/therapy , Animals , Anterior Chamber/physiology , Cell Nucleus/chemistry , Electroretinography , Erythropoietin/analysis , Gene Transfer Techniques , Intermediate Filament Proteins/genetics , Light , Membrane Glycoproteins/genetics , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Peripherins , Photoreceptor Cells, Vertebrate/physiology , Rats , Rats, Inbred Lew , Retina/chemistry , Retina/metabolism , Retina/pathology , Retinal Degeneration/etiology , Retinal Degeneration/genetics
19.
Mol Ther ; 9(2): 182-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14759802

ABSTRACT

The congenital retinal blindness known as Leber congenital amaurosis (LCA) can be caused by mutations in the RPE65 gene. RPE65 plays a critical role in the visual cycle that produces the photosensitive pigment rhodopsin. Recent evidence from human studies of LCA indicates that earlier rather than later intervention may be more likely to restore vision. We determined the impact of in utero delivery of the human RPE65 cDNA to retinal pigment epithelium cells in a murine model of LCA, the Rpe65(-/-) mouse, using a serotype 2 adeno-associated virus packaged within an AAV1 capsid (AAV2/1). Delivery of AAV2/1-CMV-hRPE65 to fetuses (embryonic day 14) resulted in efficient transduction of retinal pigment epithelium, restoration of visual function, and measurable rhodopsin. The results demonstrate AAV-mediated correction of the deficit and suggest that in utero retinal gene delivery may be a useful approach for treating a variety of blinding congenital retinal diseases.


Subject(s)
Blindness/congenital , Blindness/therapy , Disease Models, Animal , Embryo, Mammalian/metabolism , Genetic Therapy/methods , Vision, Ocular/physiology , Animals , Blindness/genetics , Blindness/physiopathology , Carrier Proteins , Dependovirus/genetics , Electroretinography , Embryo, Mammalian/embryology , Embryo, Mammalian/physiology , Embryo, Mammalian/physiopathology , Eye Proteins , Female , Genetic Vectors/genetics , Mice , Mice, Knockout , Proteins/genetics , Proteins/metabolism , Retina/embryology , Retina/metabolism , Retina/physiology , Uterus , Vision, Ocular/genetics , cis-trans-Isomerases
20.
Invest Ophthalmol Vis Sci ; 44(10): 4171-83, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14507858

ABSTRACT

PURPOSE: Mutations in RP1 are a common cause of dominant retinitis pigmentosa (RP), but the mechanism by which the identified mutations lead to photoreceptor cell death and blindness has not been determined. To investigate the function of the RP1 protein in photoreceptors and gain insight into the mechanism of disease, gene-targeting techniques were used to produce mice with a mutant Rp1 allele that mimics the truncation alleles found to cause disease. METHODS: RT-PCR was used to amplify illegitimate RP1 transcripts from lymphoblasts. Gene targeting was used to create mice with a mutant Rp1-myc allele. Confocal immunofluorescence microscopy was used to identify the location of the mutant Rp1-myc protein in photoreceptors. The structure of the photoreceptors in the resultant Rp1-myc mice was studied by light and electron microscopy. The retinal function of the mutant mice was investigated using analysis of full-field ERGs. RESULTS: Wild-type and mutant RP1 mRNA were both detected in lymphoblasts from patients with RP1 disease. Rp1-myc mice produced a truncated version of the Rp1 protein, containing the N-terminal 662 amino acids, which localized correctly to the axoneme of the photoreceptor outer segments. Mice homozygous for the mutant Rp1-myc allele underwent a rapid-onset retinal degeneration characterized by incorrectly oriented outer segment discs that failed to stack properly into outer segments. In contrast, the photoreceptors of heterozygous mice remained relatively healthy. CONCLUSIONS: The presence of mutant RP1 mRNA in lymphoblasts from patients with RP1 disease implies that the mutant message can escape nonsense-mediated mRNA decay and that a truncated RP1 protein may be produced in the retina. The truncated Rp1-myc protein appears to be nonfunctional, and not to exert a dominant negative effect in the photoreceptors of heterozygous mice. Results from homozygous Rp1-myc mice indicate that RP1 is required for the correct orientation and higher order stacking of outer segment discs.


Subject(s)
Eye Proteins/physiology , Retinitis Pigmentosa/metabolism , Rod Cell Outer Segment/physiology , Animals , Blotting, Western , Electroretinography , Gene Expression , Gene Targeting , Humans , Mice , Mice, Knockout , Mice, Mutant Strains , Microscopy, Confocal , Microtubule-Associated Proteins , RNA, Messenger/analysis , Retinitis Pigmentosa/pathology , Reverse Transcriptase Polymerase Chain Reaction , Rod Cell Outer Segment/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...