Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Ther Methods Clin Dev ; 31: 101107, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-37868206

ABSTRACT

Most inherited retinal dystrophies display progressive photoreceptor cell degeneration leading to severe visual impairment. Optogenetic reactivation of inner retinal neurons is a promising avenue to restore vision in retinas having lost their photoreceptors. Expression of optogenetic proteins in surviving ganglion cells, the retinal output, allows them to take on the lost photoreceptive function. Nonetheless, this creates an exclusively ON retina by expression of depolarizing optogenetic proteins in all classes of ganglion cells, whereas a normal retina extracts several features from the visual scene, with different ganglion cells detecting light increase (ON) and light decrease (OFF). Refinement of this therapeutic strategy should thus aim at restoring these computations. Here we used a vector that targets gene expression to a specific interneuron of the retina called the AII amacrine cell. AII amacrine cells simultaneously activate the ON pathway and inhibit the OFF pathway. We show that the optogenetic stimulation of AII amacrine cells allows restoration of both ON and OFF responses in the retina, but also mediates other types of retinal processing such as sustained and transient responses. Targeting amacrine cells with optogenetics is thus a promising avenue to restore better retinal function and visual perception in patients suffering from retinal degeneration.

2.
Sci Adv ; 8(42): eabm4295, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36260685

ABSTRACT

Accumulation of the microtubule-associated protein Tau is linked to neuronal cell death in tauopathies, but how intraneuronal Tau levels are regulated in health and disease remains unclear. Here, we show that conditional inactivation of the trafficking adaptor protein Numb in retinal ganglion cells (RGCs) increases Tau levels and leads to axonal blebbing, which is followed by neuronal cell loss in aged mice. In the TauP301S mouse model of tauopathy, conditional inactivation of Numb in RGCs and spinal motoneurons accelerates neurodegeneration, and loss of Numb in motoneurons also leads to precocious hindlimb paralysis. Conversely, overexpression of the long isoform of Numb (Numb-72) decreases intracellular Tau levels and reduces axonal blebbing in TauP301S RGCs, leading to improved electrical activity in cultured neurons and improves performance in a visually guided behavior test in vivo. These results uncover Numb as a key regulator of intracellular Tau levels and identify Numb-72 as a potential therapeutic factor for tauopathies.


Subject(s)
Tauopathies , Mice , Animals , Tauopathies/genetics , Tauopathies/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Disease Models, Animal , Retinal Ganglion Cells/metabolism , Axons/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism
3.
Mol Ther Methods Clin Dev ; 22: 15-25, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34401402

ABSTRACT

Complete congenital stationary night blindness (cCSNB) due to mutations in TRPM1, GRM6, GPR179, NYX, or leucine-rich repeat immunoglobulin-like transmembrane domain 3 (LRIT3) is an incurable inherited retinal disorder characterized by an ON-bipolar cell (ON-BC) defect. Since the disease is non-degenerative and stable, treatment could theoretically be administrated at any time in life, making it a promising target for gene therapy. Until now, adeno-associated virus (AAV)-mediated therapies lead to significant functional improvements only in newborn cCSNB mice. Here we aimed to restore protein localization and function in adult Lrit3 -/ - mice. LRIT3 localizes in the outer plexiform layer and is crucial for TRPM1 localization at the dendritic tips of ON-BCs and the electroretinogram (ERG)-b-wave. AAV2-7m8-Lrit3 intravitreal injections were performed targeting either ON-BCs, photoreceptors (PRs), or both. Protein localization of LRIT3 and TRPM1 at the rod-to-rod BC synapse, functional rescue of scotopic responses, and ON-responses detection at the ganglion cell level were achieved in a few mice when ON-BCs alone or both PRs and ON-BCs, were targeted. More importantly, a significant number of treated adult Lrit3 -/- mice revealed an ERG b-wave recovery under scotopic conditions, improved optomotor responses, and on-time ON-responses at the ganglion cell level when PRs were targeted. Functional rescue was maintained for at least 4 months after treatment.

4.
Invest Ophthalmol Vis Sci ; 62(3): 24, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33729473

ABSTRACT

Purpose: Complete congenital stationary night blindness (cCSNB) is an incurable inherited retinal disorder characterized by an ON-bipolar cell (ON-BC) defect. GRM6 mutations are the third most prevalent cause of cCSNB. The Grm6-/- mouse model mimics the human phenotype, showing no b-wave in the electroretinogram (ERG) and a loss of mGluR6 and other proteins of the same cascade at the outer plexiform layer (OPL). Our aim was to restore protein localization and function in Grm6-/- adult mice targeting specifically ON-BCs or the whole retina. Methods: Adeno-associated virus-encoding Grm6 under two different promoters (GRM6-Grm6 and CAG-Grm6) were injected intravitreally in P15 Grm6-/- mice. ERG recordings at 2 and 4 months were performed in Grm6+/+, untreated and treated Grm6-/- mice. Similarly, immunolocalization studies were performed on retinal slices before or after treatment using antibodies against mGluR6, TRPM1, GPR179, RGS7, RGS11, Gß5, and dystrophin. Results: Following treatment, mGluR6 was localized to the dendritic tips of ON-BCs when expressed with either promoter. The relocalization efficiency in mGluR6-transduced retinas at the OPL was 2.5% versus 11% when the GRM6-Grm6 and CAG-Grm6 were used, respectively. Albeit no functional rescue was seen in ERGs, relocalization of TRPM1, GPR179, and Gß5 was also noted using both constructs. The restoration of the localization of RGS7, RGS11, and dystrophin was more obvious in retinas treated with GRM6-Grm6 than in retinas treated with CAG-Grm6. Conclusions: Our findings show the potential of treating cCSNB with GRM6 mutations; however, it appears that the transduction rate must be improved to restore visual function.


Subject(s)
Dependovirus/genetics , Disease Models, Animal , Eye Diseases, Hereditary/metabolism , Gene Transfer Techniques , Genetic Diseases, X-Linked/metabolism , Myopia/metabolism , Night Blindness/metabolism , Receptors, Metabotropic Glutamate/metabolism , Retinal Bipolar Cells/metabolism , Animals , Electroretinography , Eye Diseases, Hereditary/physiopathology , Genetic Diseases, X-Linked/physiopathology , Genetic Vectors , Intravitreal Injections , Mice , Mice, Inbred C57BL , Myopia/physiopathology , Night Blindness/physiopathology , Promoter Regions, Genetic , Receptors, Metabotropic Glutamate/genetics , Retina/physiopathology , Transfection
5.
Commun Biol ; 4(1): 125, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504896

ABSTRACT

Vision restoration is an ideal medical application for optogenetics, because the eye provides direct optical access to the retina for stimulation. Optogenetic therapy could be used for diseases involving photoreceptor degeneration, such as retinitis pigmentosa or age-related macular degeneration. We describe here the selection, in non-human primates, of a specific optogenetic construct currently tested in a clinical trial. We used the microbial opsin ChrimsonR, and showed that the AAV2.7m8 vector had a higher transfection efficiency than AAV2 in retinal ganglion cells (RGCs) and that ChrimsonR fused to tdTomato (ChR-tdT) was expressed more efficiently than ChrimsonR. Light at 600 nm activated RGCs transfected with AAV2.7m8 ChR-tdT, from an irradiance of 1015 photons.cm-2.s-1. Vector doses of 5 × 1010 and 5 × 1011 vg/eye transfected up to 7000 RGCs/mm2 in the perifovea, with no significant immune reaction. We recorded RGC responses from a stimulus duration of 1 ms upwards. When using the recorded activity to decode stimulus information, we obtained an estimated visual acuity of 20/249, above the level of legal blindness (20/400). These results lay the groundwork for the ongoing clinical trial with the AAV2.7m8 - ChR-tdT vector for vision restoration in patients with retinitis pigmentosa.


Subject(s)
Optogenetics , Photic Stimulation , Retinal Degeneration/therapy , Vision, Ocular/physiology , Animals , Equipment and Supplies , Female , Humans , Macaca fascicularis , Male , Optogenetics/instrumentation , Optogenetics/methods , Pattern Recognition, Visual/physiology , Photic Stimulation/instrumentation , Photic Stimulation/methods , Primates , Retinal Degeneration/physiopathology , Retinal Degeneration/rehabilitation , Therapies, Investigational/instrumentation , Therapies, Investigational/methods
6.
Hum Mol Genet ; 28(1): 105-123, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30239717

ABSTRACT

Variations in the human Crumbs homolog-1 (CRB1) gene lead to an array of retinal dystrophies including early onset of retinitis pigmentosa (RP) and Leber congenital amaurosis (LCA) in children. To investigate the physiological roles of CRB1 and CRB2 in retinal Müller glial cells (MGCs), we analysed mouse retinas lacking both proteins in MGC. The peripheral retina showed a faster progression of dystrophy than the central retina. The central retina showed retinal folds, disruptions at the outer limiting membrane, protrusion of photoreceptor nuclei into the inner and outer segment layers and ingression of photoreceptor nuclei into the photoreceptor synaptic layer. The peripheral retina showed a complete loss of the photoreceptor synapse layer, intermingling of photoreceptor nuclei within the inner nuclear layer and ectopic photoreceptor cells in the ganglion cell layer. Electroretinography showed severe attenuation of the scotopic a-wave at 1 month of age with responses below detection levels at 3 months of age. The double knockout mouse retinas mimicked a phenotype equivalent to a clinical LCA phenotype due to loss of CRB1. Localization of CRB1 and CRB2 in non-human primate (NHP) retinas was analyzed at the ultrastructural level. We found that NHP CRB1 and CRB2 proteins localized to the subapical region adjacent to adherens junctions at the outer limiting membrane in MGC and photoreceptors. Our data suggest that loss of CRB2 in MGC aggravates the CRB1-associated RP-like phenotype towards an LCA-like phenotype.


Subject(s)
Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Retinitis Pigmentosa/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/physiology , Disease Models, Animal , Electroretinography , Ependymoglial Cells/metabolism , Ependymoglial Cells/physiology , Eye Proteins/genetics , Eye Proteins/physiology , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/physiopathology , Macaca fascicularis , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Nerve Tissue Proteins/physiology , Neuroglia/physiology , Phenotype , Photoreceptor Cells/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Retinal Dystrophies/metabolism , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/physiopathology
7.
JCI Insight ; 3(2)2018 01 25.
Article in English | MEDLINE | ID: mdl-29367457

ABSTRACT

Intraocular injection of adeno-associated viral (AAV) vectors has been an evident route for delivering gene drugs into the retina. However, gaps in our understanding of AAV transduction patterns within the anatomically unique environments of the subretinal and intravitreal space of the primate eye impeded the establishment of noninvasive and efficient gene delivery to foveal cones in the clinic. Here, we establish new vector-promoter combinations to overcome the limitations associated with AAV-mediated cone transduction in the fovea with supporting studies in mouse models, human induced pluripotent stem cell-derived organoids, postmortem human retinal explants, and living macaques. We show that an AAV9 variant provides efficient foveal cone transduction when injected into the subretinal space several millimeters away from the fovea, without detaching this delicate region. An engineered AAV2 variant provides gene delivery to foveal cones with a well-tolerated dose administered intravitreally. Both delivery modalities rely on a cone-specific promoter and result in high-level transgene expression compatible with optogenetic vision restoration. The model systems described here provide insight into the behavior of AAV vectors across species to obtain safety and efficacy needed for gene therapy in neurodegenerative disorders.


Subject(s)
Fovea Centralis/pathology , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Transduction, Genetic/methods , Vision Disorders/therapy , Animals , Cell Line , Dependovirus/genetics , Female , Fovea Centralis/diagnostic imaging , Genetic Vectors/genetics , Humans , Induced Pluripotent Stem Cells , Injections, Intraocular , Intravital Microscopy , Macaca fascicularis , Male , Mice , Models, Animal , Optogenetics/methods , Patch-Clamp Techniques , Promoter Regions, Genetic/genetics , Transgenes/genetics , Vision Disorders/genetics , Vision Disorders/pathology
8.
Methods Mol Biol ; 1715: 225-238, 2018.
Article in English | MEDLINE | ID: mdl-29188517

ABSTRACT

Adeno-associated viral vectors have become widely used in the clinic for retinal gene therapy. Thanks to AAVs impeccable safety profile and positive functional outcomes in its clinical application, interest in retinal gene therapy has increased exponentially over the past decade. Although early clinical trials have shown there is little influence of neutralizing antibodies on the performance of AAV when vector is administered into the subretinal space, recent findings suggest neutralizing antibodies may play a role when AAV is delivered via the intravitreal route. These findings highlight the importance of microenvironment on gene delivery and stress the need for a versatile assay to screen subjects for the presence of AAV-neutralizing antibodies. Measuring NAb titers against AAV prior and after gene therapy will help us better understand the impact of preexisting immunity on gene transfer, especially when the vector is administered intravitreally.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Dependovirus/immunology , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Retina/metabolism , Antibodies, Viral/blood , Antibodies, Viral/immunology , Dependovirus/genetics , HEK293 Cells , Humans , Transduction, Genetic
9.
Mol Ther ; 25(11): 2546-2560, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28807567

ABSTRACT

The majority of inherited retinal degenerations converge on the phenotype of photoreceptor cell death. Second- and third-order neurons are spared in these diseases, making it possible to restore retinal light responses using optogenetics. Viral expression of channelrhodopsin in the third-order neurons under ubiquitous promoters was previously shown to restore visual function, albeit at light intensities above illumination safety thresholds. Here, we report (to our knowledge, for the first time) activation of macaque retinas, up to 6 months post-injection, using channelrhodopsin-Ca2+-permeable channelrhodopsin (CatCh) at safe light intensities. High-level CatCh expression was achieved due to a new promoter based on the regulatory region of the gamma-synuclein gene (SNCG) allowing strong expression in ganglion cells across species. Our promoter, in combination with clinically proven adeno-associated virus 2 (AAV2), provides CatCh expression in peri-foveolar ganglion cells responding robustly to light under the illumination safety thresholds for the human eye. On the contrary, the threshold of activation and the proportion of unresponsive cells were much higher when a ubiquitous promoter (cytomegalovirus [CMV]) was used to express CatCh. The results of our study suggest that the inclusion of optimized promoters is key in the path to clinical translation of optogenetics.


Subject(s)
Channelrhodopsins/genetics , Genetic Vectors/administration & dosage , Promoter Regions, Genetic , Recovery of Function , Retinal Degeneration/therapy , Animals , Channelrhodopsins/metabolism , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Gene Expression , Genetic Therapy/methods , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Intravitreal Injections , Light , Macaca fascicularis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Optogenetics , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Transduction, Genetic , Transgenes , Vision, Ocular/physiology
10.
EMBO Mol Med ; 8(11): 1248-1264, 2016 11.
Article in English | MEDLINE | ID: mdl-27679671

ABSTRACT

Targeting the photosensitive ion channel channelrhodopsin-2 (ChR2) to the retinal circuitry downstream of photoreceptors holds promise in treating vision loss caused by retinal degeneration. However, the high intensity of blue light necessary to activate channelrhodopsin-2 exceeds the safety threshold of retinal illumination because of its strong potential to induce photochemical damage. In contrast, the damage potential of red-shifted light is vastly lower than that of blue light. Here, we show that a red-shifted channelrhodopsin (ReaChR), delivered by AAV injections in blind rd1 mice, enables restoration of light responses at the retinal, cortical, and behavioral levels, using orange light at intensities below the safety threshold for the human retina. We further show that postmortem macaque retinae infected with AAV-ReaChR can respond with spike trains to orange light at safe intensities. Finally, to directly address the question of translatability to human subjects, we demonstrate for the first time, AAV- and lentivirus-mediated optogenetic spike responses in ganglion cells of the postmortem human retina.


Subject(s)
Genetic Therapy/methods , Phototherapy/methods , Retina/physiology , Retinal Degeneration/therapy , Rhodopsin/genetics , Animals , Dependovirus/genetics , Genetic Vectors , Humans , Lentivirus/genetics , Light , Macaca , Mice , Rhodopsin/metabolism , Transduction, Genetic , Treatment Outcome
11.
Biotechnol Bioeng ; 113(12): 2712-2724, 2016 12.
Article in English | MEDLINE | ID: mdl-27259396

ABSTRACT

Recently, we described a modified AAV2 vector-AAV2-7m8-having a capsid-displayed peptide insertion of 10 amino acids with enhanced retinal transduction properties. The insertion of the peptide referred to as 7m8 is responsible for high-level gene delivery into deep layers of the retina when virus is delivered into the eye's vitreous. Here, we further characterize AAV2-7m8 mediated gene delivery to neural tissue and investigate the mechanisms by which the inserted peptide provides better transduction away from the injection site. First, in order to understand if the peptide exerts its effect on its own or in conjunction with the neighboring amino acids, we inserted the 7m8 peptide at equivalent positions on three other AAV capsids, AAV5, AAV8, and AAV9, and evaluated its effect on their infectivity. Intravitreal delivery of these peptide insertion vectors revealed that only AAV9 benefited from 7m8 insertion in the context of the retina. We then investigated AAV2-7m8 and AAV9-7m8 properties in the brain, to better evaluate the spread and efficacy of viral transduction in view of the peptide insertion. While 7m8 insertion led to higher intensity gene expression, the spread of gene expression remained unchanged compared to the parental serotypes. Our results indicate that the 7m8 peptide insertion acts by increasing efficacy of cellular entry, with little effect on the spread of viral particles in neural tissue. The effects of peptide insertion are capsid and tissue dependent, highlighting the importance of the microenvironment in gene delivery using AAV. Biotechnol. Bioeng. 2016;113: 2712-2724. © 2016 Wiley Periodicals, Inc.


Subject(s)
Capsid Proteins/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Recombinant Proteins/metabolism , Retina/virology , Transduction, Genetic/methods , Animals , Capsid Proteins/chemistry , Capsid Proteins/ultrastructure , Dependovirus/chemistry , Dependovirus/ultrastructure , Genetic Variation/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Molecular Conformation , Recombinant Proteins/genetics , Retina/physiology
12.
Mol Ther ; 23(5): 918-930, 2015 May.
Article in English | MEDLINE | ID: mdl-25619725

ABSTRACT

The promising clinical results obtained for ocular gene therapy in recent years have paved the way for gene supplementation to treat recessively inherited forms of retinal degeneration. The situation is more complex for dominant mutations, as the toxic mutant gene product must be removed. We used spliceosome-mediated RNA trans-splicing as a strategy for repairing the transcript of the rhodopsin gene, the gene most frequently mutated in autosomal dominant retinitis pigmentosa. We tested 17 different molecules targeting the pre-mRNA intron 1, by transient transfection of HEK-293T cells, with subsequent trans-splicing quantification at the transcript level. We found that the targeting of some parts of the intron promoted trans-splicing more efficiently than the targeting of other areas, and that trans-splicing rate could be increased by modifying the replacement sequence. We then developed cell lines stably expressing the rhodopsin gene, for the assessment of phenotypic criteria relevant to the pathogenesis of retinitis pigmentosa. Using this model, we showed that trans-splicing restored the correct localization of the protein to the plasma membrane. Finally, we tested our best candidate by AAV gene transfer in a mouse model of retinitis pigmentosa that expresses a mutant allele of the human rhodopsin gene, and demonstrated the feasibility of trans-splicing in vivo. This work paves the way for trans-splicing gene therapy to treat retinitis pigmentosa due to rhodopsin gene mutation and, more generally, for the treatment of genetic diseases with dominant transmission.


Subject(s)
Genetic Therapy , RNA, Messenger/genetics , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , Trans-Splicing , Animals , Binding Sites , Cell Line, Transformed , Gene Expression , Genes, Dominant , Genetic Vectors/genetics , Humans , Introns , Mice , Mice, Transgenic , Phenotype , Photoreceptor Cells/metabolism , Protein Interaction Domains and Motifs/genetics , Protein Transport , RNA Precursors/genetics , RNA Splicing , Retinitis Pigmentosa/therapy , Rhodopsin/chemistry , Rhodopsin/metabolism , Transduction, Genetic
13.
Mol Ther ; 23(1): 7-16, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25095892

ABSTRACT

Most inherited retinal dystrophies display progressive photoreceptor cell degeneration leading to severe visual impairment. Optogenetic reactivation of retinal neurons mediated by adeno-associated virus (AAV) gene therapy has the potential to restore vision regardless of patient-specific mutations. The challenge for clinical translatability is to restore a vision as close to natural vision as possible, while using a surgically safe delivery route for the fragile degenerated retina. To preserve the visual processing of the inner retina, we targeted ON bipolar cells, which are still present at late stages of disease. For safe gene delivery, we used a recently engineered AAV variant that can transduce the bipolar cells after injection into the eye's easily accessible vitreous humor. We show that AAV encoding channelrhodopsin under the ON bipolar cell-specific promoter mediates long-term gene delivery restricted to ON-bipolar cells after intravitreal administration. Channelrhodopsin expression in ON bipolar cells leads to restoration of ON and OFF responses at the retinal and cortical levels. Moreover, light-induced locomotory behavior is restored in treated blind mice. Our results support the clinical relevance of a minimally invasive AAV-mediated optogenetic therapy for visual restoration.


Subject(s)
Blindness/therapy , Dependovirus/genetics , Genetic Therapy/methods , Retinal Bipolar Cells/metabolism , Retinal Degeneration/therapy , Animals , Behavior, Animal , Blindness/genetics , Blindness/pathology , Channelrhodopsins , Female , Gene Expression , Gene Transfer Techniques , Genetic Engineering , Genetic Vectors , Intravitreal Injections , Light , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Retinal Bipolar Cells/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Visual Perception/genetics , Vitreous Body
14.
PLoS One ; 8(4): e61618, 2013.
Article in English | MEDLINE | ID: mdl-23613884

ABSTRACT

Widespread gene delivery to the retina is an important challenge for the treatment of retinal diseases, such as retinal dystrophies. We and others have recently shown that the intravenous injection of a self-complementary (sc) AAV9 vector can direct efficient cell transduction in the central nervous system, in both neonatal and adult animals. We show here that the intravenous injection of scAAV9 encoding green fluorescent protein (GFP) resulted in gene transfer to all layers of the retina in adult mice, despite the presence of a mature blood-eye barrier. Cell morphology studies and double-labeling with retinal cell-specific markers showed that GFP was expressed in retinal pigment epithelium cells, photoreceptors, bipolar cells, Müller cells and retinal ganglion cells. The cells on the inner side of the retina, including retinal ganglion cells in particular, were transduced with the highest efficiency. Quantification of the cell population co-expressing GFP and Brn-3a showed that 45% of the retinal ganglion cells were efficiently transduced after intravenous scAAV9-GFP injection in adult mice. This study provides the first demonstration that a single intravenous scAAV9 injection can deliver transgenes to the retinas of both eyes in adult mice, suggesting that this vector serotype is able to cross mature blood-eye barriers. This intravascular gene transfer approach, by eliminating the potential invasiveness of ocular surgery, could constitute an alternative when fragility of the retina precludes subretinal or intravitreal injections of viral vectors, opening up new possibilities for gene therapy for retinal diseases.


Subject(s)
Dependovirus/genetics , Retina/metabolism , Transduction, Genetic/methods , Animals , Blood/virology , Female , Genetic Vectors/genetics , Injections, Intravenous , Mice , Mice, Inbred C57BL , Optic Nerve/metabolism , Retina/cytology , Retina/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...