Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Article in English | MEDLINE | ID: mdl-37848250

ABSTRACT

Rodent models of retinal degeneration are essential for the development of therapeutic strategies. In addition to living animal models, we here also discuss models based on rodent cell cultures, such as purified retinal ganglion cells and retinal explants. These ex vivo models extend the possibilities for investigating pathological mechanisms and assessing the neuroprotective effect of pharmacological agents by eliminating questions on drug pharmacokinetics and bioavailability. The number of living rodent models has greatly increased with the possibilities to achieve transgenic modifications in animals for knocking in and out genes and mutations. The Cre-lox system has further enabled investigators to target specific genes or mutations in specific cells at specific stages. However, chemically or physically induced models can provide alternatives to such targeted gene modifications. The increased diversity of rodent models has widened our possibility to address most ocular pathologies for providing initial proof of concept of innovative therapeutic strategies.

2.
Ophthalmol Sci ; 3(4): 100316, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37274010

ABSTRACT

Objective: To describe adaptive optics flood illumination ophthalmoscopy (AO-FIO) of the photoreceptor layer in normal nonhuman primates (NHPs) and in the case of a short-term induced retinal detachment (RD). Design: Longitudinal fundamental research study. Subjects: Four NHPs were used to image normal retinae with AO-FIO (in comparison with 4 healthy humans); 2 NHPs were used to assess the effects of RD. Intervention: The photoreceptor layer (cone mosaic metrics, including cone density, cone spacing, and cone regularity) was followed with AO-FIO imaging (rtx1, Imagine Eyes) during a surgically induced RD in 2 NHPs using a vehicle solution containing dimethyl sulfoxide, classically used as a chemical solvent. We also performed functional testing of the retina (full-field and multifocal electroretinogram [ERG]). Main Outcome Measures: Correlation of cone mosaic metrics (cone density, spacing, and regularity) between normal retinae of NHPs and humans, and cone metrics, power spectrum, and ERG wave amplitudes after RD. Results: Imaging features were very similar in terms of cone reflectivity, cell density, regularity, and spacing values, showing strong positive correlations between NHPs and humans. After RD, AO-FIO revealed several alterations of the cone mosaic slowly recovering during the 3 months after the reattachment, which were not detected functionally by ERG. Conclusions: These results demonstrate by in vivo AO-FIO imaging the transient structural changes of photoreceptors after an RD in the primate retina. They also provide an interesting illustration of the AO-FIO potential for investigating photoreceptor toxicity during preclinical studies in NHPs with a high translatability to human studies. Financial Disclosures: Proprietary or commercial disclosure may be found after the references.

3.
Mol Ther ; 31(7): 2005-2013, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37016579

ABSTRACT

Lenadogene nolparvovec (GS010) was developed to treat a point mutation in mitochondrial ND4 that causes Leber hereditary optic neuropathy. GS010 delivers human cDNA encoding wild-type ND4 packaged into an rAAV2/2 vector that transduces retinal ganglion cells, to induce allotopic expression of hybrid mitochondrial ND4. GS010 clinical trials improved best-corrected visual acuity (BCVA) up to 5 years after treatment. Interestingly, unilateral treatment improved BCVA bilaterally. Subsequent studies revealed GS010 DNA in visual tissues contralateral to the injected eye, suggesting migration. Here we tested whether unilateral intraocular pressure (IOP) elevation could influence the transfer of viral ND4 RNA in contralateral tissues after GS010 delivery to the IOP-elevated eye and probed a potential mechanism mediating translocation in mice. We found IOP elevation enhanced viral ND4 RNA transcripts in contralateral visual tissues, including retinas. Using conditional transgenic mice, we depleted astrocytic gap junction connexin 43 (Cx43), required for distant redistribution of metabolic resources between astrocytes during stress. After unilateral IOP elevation and GS010 injection, Cx43 knockdown eradicated ND4 RNA transcript detection in contralateral retinal tissues, while transcript was still detectable in optic nerves. Overall, our study indicates long-range migration of GS010 product to contralateral visual tissues is enhanced by Cx43-linked astrocyte networks.


Subject(s)
Astrocytes , Connexin 43 , Mice , Humans , Animals , Astrocytes/metabolism , Connexin 43/genetics , Connexin 43/metabolism , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Dependovirus/genetics , Dependovirus/metabolism , Genetic Vectors , Genetic Therapy , Mice, Transgenic , RNA , DNA, Mitochondrial/genetics
4.
J Adv Res ; 45: 141-155, 2023 03.
Article in English | MEDLINE | ID: mdl-35623612

ABSTRACT

INTRODUCTION: Corneal blindness due to scarring is treated with corneal transplantation. However, a global problem is the donor material shortage. Preclinical and clinical studies have shown that cell-based therapy using corneal stromal stem cells (CSSCs) suppresses corneal scarring, potentially mediated by specific microRNAs transported in extracellular vesicles (EVs). However, not every CSSC batch from donors achieves similar anti-scarring effects. OBJECTIVES: To examine miRNA profiles in EVs from human CSSCs showing "healing" versus "non-healing" effects on corneal scarring and to design a tool to select CSSCs with strong healing potency for clinical applications. METHODS: Small RNAs from CSSC-EVs were extracted for Nanostring nCounter Human miRNA v3 assay. MicroRNAs expressed > 20 folds in "healing" EVs (P < 0.05) were subject to enriched gene ontology (GO) term analysis. MiRNA groups with predictive regulation on inflammatory and fibrotic signalling were studied by mimic transfection to (1) mouse macrophages (RAW264.7) for M1 phenotype assay; (2) human corneal keratocytes for cytokine-induced fibrosis, and (3) human CSSCs for corneal scar prevention in vivo. The expression of miR-29a was screened in additional CSSC batches and the anti-scarring effect of cells was validated in mouse corneal wounds. RESULTS: Twenty-one miRNAs were significantly expressed in "healing" CSSC-EVs and 9 miRNA groups were predicted to associate with inflammatory and fibrotic responses, and tissue regeneration (P <10-6). Overexpression of miR-29a and 381-5p significantly prevented M1 phenotype transition in RAW264.7 cells after lipopolysaccharide treatment, suppressed transforming growth factor ß1-induced fibrosis marker expression in keratocytes, and reduced scarring after corneal injury. High miR-29a expression in EV fractions distinguished human CSSCs with strong healing potency, which inhibited corneal scarring in vivo. CONCLUSION: We characterized the anti-inflammatory and fibrotic roles of miR-29a and 381-5p in CSSCs, contributing to scar prevention. MiR-29a expression in EVs distinguished CSSCs with anti-scarring quality, identifying good quality cells for a scarless corneal healing.


Subject(s)
Corneal Injuries , MicroRNAs , Humans , Animals , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Corneal Injuries/therapy , Stem Cells/metabolism , Cicatrix , Fibrosis , Cell- and Tissue-Based Therapy
5.
J Neural Eng ; 19(5)2022 10 14.
Article in English | MEDLINE | ID: mdl-36174540

ABSTRACT

Objective. Subretinal prostheses electrically stimulate the residual inner retinal neurons to partially restore vision. We investigated the changes in neurosensory macular structures and it is thickness associated with subretinal implantation in geographic atrophy (GA) secondary to age-related macular degeneration (AMD).Approach. Using optical coherence tomography, changes in distance between electrodes and retinal inner nuclear layer (INL) as well as alterations in thickness of retinal layers were measured over time above and near the subretinal chip implanted within the atrophic area. Retinal thickness (RT) was quantified across the implant surface and edges as well as outside the implant zone to compare with the natural macular changes following subretinal surgery, and the natural course of dry AMD.Main results. GA was defined based on complete retinal pigment epithelium and outer retinal atrophy (cRORA). Based on the analysis of three patients with subretinal implantation, we found that the distance between the implant and the target cells was stable over the long-term follow-up. Total RT above the implant decreased on average, by 39 ± 12µm during 3 months post-implantation, but no significant changes were observed after that, up to 36 months of the follow-up. RT also changed near the temporal entry point areas outside the implantation zone following the surgical trauma of retinal detachment. There was no change in the macula cRORA nasal to the implanted zone, where there was no surgical trauma or manipulation.Significance. The surgical delivery of the photovoltaic subretinal implant causes minor RT changes that settle after 3 months, and then remain stable over long-term with no adverse structural or functional effects. Distance between the implant and the INL remains stable up to 36 months of the follow-up.


Subject(s)
Geographic Atrophy , Macular Degeneration , Geographic Atrophy/etiology , Geographic Atrophy/surgery , Humans , Macular Degeneration/complications , Macular Degeneration/surgery , Prostheses and Implants , Tomography, Optical Coherence/methods , Visual Acuity
6.
Mol Ther Methods Clin Dev ; 23: 307-318, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34729378

ABSTRACT

Lenadogene nolparvovec (Lumevoq) gene therapy was developed to treat Leber hereditary optic neuropathy (LHON) caused by the m.11778G > A in MT-ND4 that affects complex I of the mitochondrial respiratory chain. Lenadogene nolparvovec is a replication-defective, single-stranded DNA recombinant adeno-associated virus vector 2 serotype 2, containing a codon-optimized complementary DNA encoding the human wild-type MT-ND4 subunit protein. Lenadogene nolparvovec was administered by unilateral intravitreal injection in MT-ND4 LHON patients in two randomized, double-masked, and sham-controlled phase III clinical trials (REVERSE and RESCUE), resulting in bilateral improvement of visual acuity. These and other earlier results suggest that lenadogene nolparvovec may travel from the treated to the untreated eye. To investigate this possibility further, lenadogene nolparvovec was unilaterally injected into the vitreous body of the right eye of healthy, nonhuman primates. Viral vector DNA was quantifiable in all eye and optic nerve tissues of the injected eye and was detected at lower levels in some tissues of the contralateral, noninjected eye, and optic projections, at 3 and 6 months after injection. The results suggest that lenadogene nolparvovec transfers from the injected to the noninjected eye, thus providing a potential explanation for the bilateral improvement of visual function observed in the LHON patients.

7.
Transl Vis Sci Technol ; 9(7): 2, 2020 06.
Article in English | MEDLINE | ID: mdl-32832209

ABSTRACT

Major advances in the study of inherited retinal diseases (IRDs) have placed efforts to develop treatments for these blinding conditions at the forefront of the emerging field of precision medicine. As a result, the growth of clinical trials for IRDs has increased rapidly over the past decade and is expected to further accelerate as more therapeutic possibilities emerge and qualified participants are identified. Although guided by established principles, these specialized trials, requiring analysis of novel outcome measures and endpoints in small patient populations, present multiple challenges relative to study design and ethical considerations. This position paper reviews recent accomplishments and existing challenges in clinical trials for IRDs and presents a set of recommendations aimed at rapidly advancing future progress. The goal is to stimulate discussions among researchers, funding agencies, industry, and policy makers that will further the design, conduct, and analysis of clinical trials needed to accelerate the approval of effective treatments for IRDs, while promoting advocacy and ensuring patient safety.


Subject(s)
Retinal Diseases , Humans , Precision Medicine , Retina , Retinal Diseases/drug therapy
8.
Ophthalmology ; 127(8): 1097-1104, 2020 08.
Article in English | MEDLINE | ID: mdl-32249038

ABSTRACT

PURPOSE: Loss of photoreceptors in atrophic age-related macular degeneration results in severe visual impairment, although some peripheral vision is retained. To restore central vision without compromising the residual peripheral field, we developed a wireless photovoltaic retinal implant (PRIMA; Pixium Vision, Paris, France) in which pixels convert images projected from video glasses using near-infrared light into electric current to stimulate the nearby inner retinal neurons. DESIGN: We carried out a first-in-human clinical trial to test the safety and efficacy of the prosthesis in patients with geographic atrophy (ClinicalTrials.gov identifier, NCT03333954). PARTICIPANTS: Five patients with geographic atrophy zone of at least 3 optic disc diameters, no foveal light perception, and best-corrected visual acuity of 20/400 to 20/1000 in the worse-seeing study eye. METHODS: The 2-mm wide, 30-µm thick chip, containing 378 pixels (each 100 µm in diameter), was implanted subretinally in the area of atrophy (absolute scotoma). MAIN OUTCOME MEASURES: Anatomic outcomes were assessed with fundus photography and OCT for up to 12 months of follow-up. Prosthetic vision was assessed by mapping light perception, bar orientation, letter recognition, and Landolt C acuity. RESULTS: In all patients, the prosthesis was implanted successfully under the macula, although in 2 patients, it was implanted in unintended locations: within the choroid and off center by 2 mm. All 5 patients could perceive white-yellow prosthetic visual patterns with adjustable brightness in the previous scotomata. The 3 with optimal placement of the implant demonstrated prosthetic acuity of 20/460 to 20/550, and the patient with the off-center implant demonstrated 20/800 acuity. Residual natural acuity did not decrease after implantation in any patient. CONCLUSIONS: Implantation of the PRIMA did not decrease the residual natural acuity, and it restored visual sensitivity in the former scotoma in each of the 5 patients. In 3 patients with the proper placement of the chip, prosthetic visual acuity was only 10% to 30% less than the level expected from the pixel pitch (20/420). Therefore, the use of optical or electronic magnification in the glasses as well as smaller pixels in future implants may improve visual acuity even further.


Subject(s)
Electrodes, Implanted , Macula Lutea/pathology , Macular Degeneration/physiopathology , Recovery of Function/physiology , Visual Perception/physiology , Visual Prosthesis , Feasibility Studies , Female , Humans , Macular Degeneration/diagnosis , Male , Middle Aged , Tomography, Optical Coherence
10.
Sci Rep ; 8(1): 1968, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29386551

ABSTRACT

Usher syndrome type 1 (USH1) is a major cause of inherited deafness and blindness in humans. The eye disorder is often referred to as retinitis pigmentosa, which is characterized by a secondary cone degeneration following the rod loss. The development of treatments to prevent retinal degeneration has been hampered by the lack of clear evidence for retinal degeneration in mutant mice deficient for the Ush1 genes, which instead faithfully mimic the hearing deficit. We show that, under normal housing conditions, Ush1g-/- and Ush1c-/- albino mice have dysfunctional cone photoreceptors whereas pigmented knockout animals have normal photoreceptors. The key involvement of oxidative stress in photoreceptor apoptosis and the ensued retinal gliosis were further confirmed by their prevention when the mutant mice are reared under darkness and/or supplemented with antioxidants. The primary degeneration of cone photoreceptors contrasts with the typical forms of retinitis pigmentosa. Altogether, we propose that oxidative stress probably accounts for the high clinical heterogeneity among USH1 siblings, which also unveils potential targets for blindness prevention.


Subject(s)
Antioxidants/therapeutic use , Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/drug therapy , Retinal Degeneration/prevention & control , Animals , Antioxidants/pharmacology , Apoptosis , Cell Cycle Proteins , Cytoskeletal Proteins , Darkness , Diet , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Housing, Animal , Mice, Inbred BALB C , Mice, Inbred C57BL , Opsins/metabolism , Phenotype , Retinal Cone Photoreceptor Cells/drug effects , Retinal Degeneration/pathology , Taurine/administration & dosage
12.
Nat Commun ; 6: 8666, 2015 Oct 21.
Article in English | MEDLINE | ID: mdl-26487268

ABSTRACT

Ciliopathies are a large group of clinically and genetically heterogeneous disorders caused by defects in primary cilia. Here we identified mutations in TRAF3IP1 (TNF Receptor-Associated Factor Interacting Protein 1) in eight patients from five families with nephronophthisis (NPH) and retinal degeneration, two of the most common manifestations of ciliopathies. TRAF3IP1 encodes IFT54, a subunit of the IFT-B complex required for ciliogenesis. The identified mutations result in mild ciliary defects in patients but also reveal an unexpected role of IFT54 as a negative regulator of microtubule stability via MAP4 (microtubule-associated protein 4). Microtubule defects are associated with altered epithelialization/polarity in renal cells and with pronephric cysts and microphthalmia in zebrafish embryos. Our findings highlight the regulation of cytoplasmic microtubule dynamics as a role of the IFT54 protein beyond the cilium, contributing to the development of NPH-related ciliopathies.


Subject(s)
Carrier Proteins/genetics , Kidney Diseases, Cystic/genetics , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mutation , Retinal Degeneration/genetics , Zebrafish Proteins/genetics , Animals , Blotting, Western , Carrier Proteins/metabolism , Cell Polarity/genetics , Circular Dichroism , Embryo, Nonmammalian , Female , Fluorescent Antibody Technique , Gene Knockout Techniques , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Immunoprecipitation , Kidney Diseases, Cystic/metabolism , Male , Microphthalmos/genetics , Pedigree , Retinal Degeneration/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish , Zebrafish Proteins/metabolism
15.
Int J Artif Organs ; 36(9): 612-9, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23918262

ABSTRACT

Retinal prostheses are being developed to restore vision in blind patients with photoreceptor degeneration. Electrodes arrays were subretinally implanted in transgenic P23H rats with their photoreceptors degenerated. Electrical stability of the implants was evaluated by long-term monitoring of their impedance changes. Electrode impedances were found to increase by two log units over a three weeks period whereas no impedance increase was noted when the implants were located in the vitreous. In case of hemorrhage or major fibrous reactions, the impedance continued to increase steadily. After explantation, it recovered its initial value indicating no deterioration of the implant. Although the glial cell layer at the surface of the subretinal space was slightly larger, no major glial reaction was seen in direct contact to the implant. These results indicate that no functional testing should be considered before at least three weeks post implantation.


Subject(s)
Prosthesis Implantation/methods , Retina/surgery , Visual Prosthesis/adverse effects , Animals , Electric Impedance , Microelectrodes , Prosthesis Implantation/adverse effects , Rats
16.
Front Neurosci ; 6: 168, 2012.
Article in English | MEDLINE | ID: mdl-23189036

ABSTRACT

Retinal prostheses, which restore partial vision to patients blinded by outer retinal degeneration, are currently in clinical trial. The Argus II retinal prosthesis system was recently awarded CE approval for commercial use in Europe. While retinal prosthesis users have achieved remarkable visual improvement to the point of reading letters and short sentences, the reading process is still fairly cumbersome. This study investigates the possibility of using an epiretinal prosthesis to stimulate visual braille as a sensory substitution for reading written letters and words. The Argus II retinal prosthesis system, used in this study, includes a 10 × 6 electrode array implanted epiretinally, a tiny video camera mounted on a pair of glasses, and a wearable computer that processes the video and determines the stimulation current of each electrode in real time. In the braille reading system, individual letters are created by a subset of dots from a 3 by 2 array of six dots. For the visual braille experiment, a grid of six electrodes was chosen out of the 10 × 6 Argus II array. Groups of these electrodes were then directly stimulated (bypassing the camera) to create visual percepts of individual braille letters. Experiments were performed in a single subject. Single letters were stimulated in an alternative forced choice (AFC) paradigm, and short 2-4-letter words were stimulated (one letter at a time) in an open-choice reading paradigm. The subject correctly identified 89% of single letters, 80% of 2-letter, 60% of 3-letter, and 70% of 4-letter words. This work suggests that text can successfully be stimulated and read as visual braille in retinal prosthesis patients.

17.
J Neural Eng ; 9(6): 066004, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23075696

ABSTRACT

Objective. Accurate modeling of retinal information processing remains a major challenge in retinal physiology with applications in visual rehabilitation and prosthetics. Most of the current artificial retinas are fed with static frame-based information, losing thereby the fundamental asynchronous features of biological vision. The objective of this work is to reproduce the spatial and temporal properties of the majority of ganglion cell (GC) types in the mammalian retina. Approach. Here, we combined an asynchronous event-based light sensor with a model pulling nonlinear subunits to reproduce the parallel filtering and temporal coding occurring in the retina. We fitted our model to physiological data and were able to reconstruct the spatio-temporal responses of the majority of GC types previously described in the mammalian retina (Roska et al 2006 J. Neurophysiol. 95 3810-22). Main results. Fitting of the temporal and spatial components of the response was achieved with high coefficients of determination (median R(2) = 0.972 and R(2) = 0.903, respectively). Our model provides an accurate temporal precision with a reliability of only few milliseconds-peak of the distribution at 5 ms-similar to biological retinas (Berry et al 1997 Proc. Natl Acad. Sci. USA 94 5411-16; Gollisch and Meister 2008 Science 319 1108-11). The spiking statistics of the model also followed physiological measurements (Fano factor: 0.331). Significance. This new asynchronous retinal model therefore opens new perspectives in the development of artificial visual systems and visual prosthetic devices.


Subject(s)
Eye, Artificial , Models, Neurological , Retina/physiology , Algorithms , Animals , Computer Graphics , Computer Simulation , Electronics , Light , Mammals , Nonlinear Dynamics , Patch-Clamp Techniques , Photic Stimulation , Retinal Ganglion Cells/physiology
18.
PLoS One ; 7(6): e37873, 2012.
Article in English | MEDLINE | ID: mdl-22675496

ABSTRACT

Glaucoma, the most common cause of irreversible blindness, is a neuropathy commonly initiated by pathological ocular hypertension due to unknown mechanisms of trabecular meshwork degeneration. Current antiglaucoma therapy does not target the causal trabecular pathology, which may explain why treatment failure is often observed. Here we show that the chemokine CXCL12, its truncated form SDF-1(5-67), and the receptors CXCR4 and CXCR3 are expressed in human glaucomatous trabecular tissue and a human trabecular cell line. SDF-1(5-67) is produced under the control of matrix metallo-proteinases, TNF-α, and TGF-ß2, factors known to be involved in glaucoma. CXCL12 protects in vitro trabecular cells from apoptotic death via CXCR4 whereas SDF-1(5-67) induces apoptosis through CXCR3 and caspase activation. Ocular administration of SDF-1(5-67) in the rat increases intraocular pressure. In contrast, administration of a selective CXCR3 antagonist in a rat model of ocular hypertension decreases intraocular pressure, prevents retinal neurodegeneration, and preserves visual function. The protective effect of CXCR3 antagonism is related to restoration of the trabecular function. These data demonstrate that proteolytic cleavage of CXCL12 is involved in trabecular pathophysiology, and that local administration of a selective CXCR3 antagonist may be a beneficial therapeutic strategy for treating ocular hypertension and subsequent retinal degeneration.


Subject(s)
Chemokine CXCL12/pharmacology , Ocular Hypertension/complications , Ocular Hypertension/physiopathology , Receptors, CXCR3/antagonists & inhibitors , Retinal Degeneration/complications , Retinal Degeneration/prevention & control , Trabecular Meshwork/physiopathology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line , Cytoprotection/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , Glaucoma/complications , Glaucoma/metabolism , Glaucoma/pathology , Glaucoma/physiopathology , Humans , Intraocular Pressure/drug effects , Male , Rats , Rats, Long-Evans , Receptors, CXCR3/metabolism , Receptors, CXCR4/metabolism , Retinal Degeneration/physiopathology , Stress, Physiological/drug effects , Trabecular Meshwork/drug effects , Trabecular Meshwork/pathology , Vision, Ocular/drug effects
19.
Science ; 329(5990): 413-7, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20576849

ABSTRACT

Retinitis pigmentosa refers to a diverse group of hereditary diseases that lead to incurable blindness, affecting two million people worldwide. As a common pathology, rod photoreceptors die early, whereas light-insensitive, morphologically altered cone photoreceptors persist longer. It is unknown if these cones are accessible for therapeutic intervention. Here, we show that expression of archaebacterial halorhodopsin in light-insensitive cones can substitute for the native phototransduction cascade and restore light sensitivity in mouse models of retinitis pigmentosa. Resensitized photoreceptors activate all retinal cone pathways, drive sophisticated retinal circuit functions (including directional selectivity), activate cortical circuits, and mediate visually guided behaviors. Using human ex vivo retinas, we show that halorhodopsin can reactivate light-insensitive human photoreceptors. Finally, we identified blind patients with persisting, light-insensitive cones for potential halorhodopsin-based therapy.


Subject(s)
Genetic Therapy , Halorhodopsins/genetics , Halorhodopsins/metabolism , Retinal Cone Photoreceptor Cells/physiology , Retinitis Pigmentosa/therapy , Animals , Dependovirus/genetics , Disease Models, Animal , Evoked Potentials, Visual , Genetic Vectors , Halobacteriaceae/genetics , Humans , Light , Mice , Mice, Knockout , Promoter Regions, Genetic , Retina/physiology , Retinal Ganglion Cells/physiology , Retinitis Pigmentosa/physiopathology , Tissue Culture Techniques , Transfection , Vision, Ocular , Visual Pathways/physiology
20.
Mol Cell Neurosci ; 43(4): 414-21, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20132888

ABSTRACT

The anti-epileptic drug vigabatrin induces an irreversible constriction of the visual field, but is still widely used to treat infantile spasms and some forms of epilepsy. We recently reported that vigabatrin-induced cone damage is due to a taurine deficiency. However, optic atrophy and thus retinal ganglion cell degeneration was also reported in children treated for infantile spasms. We here show in neonatal rats treated from postnatal days 4 to 29 that the vigabatrin treatment triggers not only cone photoreceptor damage, disorganisation of the photoreceptor layer and gliosis but also retinal ganglion cell loss. Furthermore, we demonstrate in these neonatal rats that taurine supplementation partially prevents these retinal lesions and in particular the retinal ganglion cell loss. These results provide the first evidence of retinal ganglion cell neuroprotection by taurine. They further confirm that taurine supplementation should be administered with the vigabatrin treatment for infantile spasms or epilepsy.


Subject(s)
Cell Death/drug effects , Optic Atrophy/chemically induced , Photoreceptor Cells/pathology , Retinal Ganglion Cells/pathology , Taurine/deficiency , Vigabatrin/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Anticonvulsants/pharmacology , Cell Count , Electroretinography , Fluorescent Antibody Technique , Neuroprotective Agents/administration & dosage , Optic Atrophy/pathology , Photoreceptor Cells/drug effects , Rats , Rats, Wistar , Retinal Ganglion Cells/drug effects , Taurine/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...