Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Mol Ther Methods Clin Dev ; 28: 396-411, 2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36910588

ABSTRACT

Gene therapy of Usher syndrome type 1B (USH1B) due to mutations in the large Myosin VIIA (MYO7A) gene is limited by the packaging capacity of adeno-associated viral (AAV) vectors. To overcome this, we have previously developed dual AAV8 vectors which encode human MYO7A (dual AAV8.MYO7A). Here we show that subretinal administration of 1.37E+9 to 1.37E+10 genome copies of a good-manufacturing-practice-like lot of dual AAV8.MYO7A improves the retinal defects of a mouse model of USH1B. The same lot was used in non-human primates at doses 1.6× and 4.3× the highest dose proposed for the clinical trial which was based on mouse efficacy data. Long-lasting alterations in retinal function and morphology were observed following subretinal administration of dual AAV8.MYO7A at the high dose. These findings were modest and improved over time in the low-dose group, as also observed in other studies involving the use of AAV8 in non-human primates and humans. Biodistribution and shedding studies confirmed the presence of vector DNA mainly in the visual pathway. Accordingly, we detected human MYO7A mRNA expression predominantly in the retina. Overall, these studies pave the way for the clinical translation of subretinal administration of dual AAV vectors in USH1B subjects.

2.
Nat Commun ; 13(1): 1963, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35414130

ABSTRACT

Challenges to the widespread application of gene therapy with adeno-associated viral (AAV) vectors include dominant conditions due to gain-of-function mutations which require allele-specific knockout, as well as long-term transgene expression from proliferating tissues, which is hampered by AAV DNA episomal status. To overcome these challenges, we used CRISPR/Cas9-mediated homology-independent targeted integration (HITI) in retina and liver as paradigmatic target tissues. We show that AAV-HITI targets photoreceptors of both mouse and pig retina, and this results in significant improvements to retinal morphology and function in mice with autosomal dominant retinitis pigmentosa. In addition, we show that neonatal systemic AAV-HITI delivery achieves stable liver transgene expression and phenotypic improvement in a mouse model of a severe lysosomal storage disease. We also show that HITI applications predominantly result in on-target editing. These results lay the groundwork for the application of AAV-HITI for the treatment of diseases affecting various organs.


Subject(s)
Dependovirus , Gene Editing , Animals , CRISPR-Cas Systems , Dependovirus/genetics , Gene Editing/methods , Genetic Vectors/genetics , Liver , Mice , Retina/metabolism , Swine
3.
Mol Ther Methods Clin Dev ; 23: 448-459, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34786437

ABSTRACT

Split intein-mediated protein trans-splicing expands AAV transfer capacity, thus overcoming the limited AAV cargo. However, non-mammalian inteins persist as trans-splicing by-products, and this could raise safety concerns for AAV intein clinical applications. In this study, we tested the ability of several degrons to selectively decrease levels of inteins after protein trans-splicing and found that a version of E. coli dihydrofolate reductase, which we have shortened to better fit into the AAV vector, is the most effective. We show that subretinal administration of AAV intein armed with this short degron is both safe and effective in a mouse model of Stargardt disease (STGD1), which is the most common form of inherited macular degeneration in humans. This supports the use of optimized AAV intein for gene therapy of both STGD1 and other conditions that require transfer of large genes.

4.
Am J Hum Genet ; 108(2): 295-308, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33508235

ABSTRACT

Retinitis pigmentosa (RP) is a group of progressive retinal degenerations of mostly monogenic inheritance, which cause blindness in about 1:3,500 individuals worldwide. Heterozygous variants in the rhodopsin (RHO) gene are the most common cause of autosomal dominant RP (adRP). Among these, missense variants at C-terminal proline 347, such as p.Pro347Ser, cause severe adRP recurrently in European affected individuals. Here, for the first time, we use CRISPR/Cas9 to selectively target the p.Pro347Ser variant while preserving the wild-type RHO allele in vitro and in a mouse model of adRP. Detailed in vitro, genomic, and biochemical characterization of the rhodopsin C-terminal editing demonstrates a safe downregulation of p.Pro347Ser expression leading to partial recovery of photoreceptor function in a transgenic mouse model treated with adeno-associated viral vectors. This study supports the safety and efficacy of CRISPR/Cas9-mediated allele-specific editing and paves the way for a permanent and precise correction of heterozygous variants in dominantly inherited retinal diseases.


Subject(s)
Gene Editing , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Rhodopsin/genetics , Alleles , Animals , CRISPR-Cas Systems , Cell Line , Dependovirus/genetics , Disease Models, Animal , Electroretinography , Genetic Therapy , Humans , INDEL Mutation , Mice , Mice, Transgenic , Mutation, Missense , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Retina/physiopathology , Rhodopsin/metabolism
5.
Sci Transl Med ; 11(492)2019 05 15.
Article in English | MEDLINE | ID: mdl-31092694

ABSTRACT

Retinal gene therapy with adeno-associated viral (AAV) vectors holds promises for treating inherited and noninherited diseases of the eye. Although clinical data suggest that retinal gene therapy is safe and effective, delivery of large genes is hindered by the limited AAV cargo capacity. Protein trans-splicing mediated by split inteins is used by single-cell organisms to reconstitute proteins. Here, we show that delivery of multiple AAV vectors each encoding one of the fragments of target proteins flanked by short split inteins results in protein trans-splicing and full-length protein reconstitution in the retina of mice and pigs and in human retinal organoids. The reconstitution of large therapeutic proteins using this approach improved the phenotype of two mouse models of inherited retinal diseases. Our data support the use of split intein-mediated protein trans-splicing in combination with AAV subretinal delivery for gene therapy of inherited blindness due to mutations in large genes.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Green Fluorescent Proteins/genetics , Inteins , Retina/virology , Trans-Splicing/genetics , Animals , Genetic Vectors/administration & dosage , Genetic Vectors/metabolism , Green Fluorescent Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Organoids/ultrastructure , Organoids/virology , Phenotype , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/virology , Swine
6.
Hum Gene Ther ; 29(8): 886-901, 2018 08.
Article in English | MEDLINE | ID: mdl-29641320

ABSTRACT

Retinal gene therapy based on adeno-associated viral (AAV) vectors is safe and efficient in humans. The low intrinsic DNA transfer capacity of AAV has been expanded by dual vectors where a large expression cassette is split in two halves independently packaged in two AAV vectors. Dual AAV transduction efficiency, however, is greatly reduced compared to that obtained with a single vector. As AAV intracellular trafficking and processing are negatively affected by phosphorylation, this study set to identify kinase inhibitors that can increase dual AAV vector transduction. By high-throughput screening of a kinase inhibitors library, three compounds were identified that increase AAV transduction in vitro, one of which has a higher effect on dual than on single AAV vectors. Importantly, the transduction enhancement is exerted on various AAV serotypes and is not transgene dependent. As kinase inhibitors are promiscuous, siRNA-mediated silencing of targeted kinases was performed, and AURKA and B, PLK1, and PTK2 were among those involved in the increase of AAV transduction levels. The study shows that kinase inhibitor administration reduces AAV serotype 2 (AAV2) capsid phosphorylation and increases the activity of DNA-repair pathways involved in AAV DNA processing. Importantly, the kinase inhibitor PF-00562271 improves dual AAV8 transduction in photoreceptors following sub-retinal delivery in mice. The study identifies kinase inhibitors that increase dual and single AAV transduction by modulating AAV entry and post-entry steps.


Subject(s)
Genetic Therapy , Genetic Vectors/drug effects , Protein Kinase Inhibitors/administration & dosage , Retina/metabolism , Transduction, Genetic , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Dependovirus/genetics , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation/drug effects , Genetic Vectors/therapeutic use , High-Throughput Screening Assays , Humans , Mice , Photoreceptor Cells/drug effects , Photoreceptor Cells/virology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Retina/pathology , Retina/virology , Polo-Like Kinase 1
7.
Hum Mol Genet ; 24(23): 6811-25, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26420842

ABSTRACT

Stargardt disease (STGD1) due to mutations in the large ABCA4 gene is the most common inherited macular degeneration in humans. We have shown that dual adeno-associated viral (AAV) vectors effectively transfer ABCA4 to the retina of Abca4-/- mice. However, they express both lower levels of transgene compared with a single AAV and truncated proteins. To increase productive dual AAV concatemerization, which would overcome these limitations, we have explored the use of either various regions of homology or heterologous inverted terminal repeats (ITR). In addition, we tested the ability of various degradation signals to decrease the expression of truncated proteins. We found the highest levels of transgene expression using regions of homology based on either alkaline phosphatase or the F1 phage (AK). The use of heterologous ITR does not decrease the levels of truncated proteins relative to full-length ABCA4 and impairs AAV vector production. Conversely, the inclusion of the CL1 degradation signal results in the selective degradation of truncated proteins from the 5'-half without affecting full-length protein production. Therefore, we developed dual AAV hybrid ABCA4 vectors including homologous ITR2, the photoreceptor-specific G protein-coupled receptor kinase 1 promoter, the AK region of homology and the CL1 degradation signal. We show that upon subretinal administration these vectors are both safe in pigs and effective in Abca4-/- mice. Our data support the use of improved dual AAV vectors for gene therapy of STGD1.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors , Macular Degeneration/congenital , Administration, Ophthalmic , Animals , Disease Models, Animal , Female , Genetic Vectors/administration & dosage , HEK293 Cells , Humans , Macular Degeneration/genetics , Macular Degeneration/therapy , Mice , Retina/metabolism , Stargardt Disease , Swine , Terminal Repeat Sequences , Transgenes
8.
EMBO Mol Med ; 6(2): 194-211, 2014 02.
Article in English | MEDLINE | ID: mdl-24150896

ABSTRACT

Retinal gene therapy with adeno-associated viral (AAV) vectors is safe and effective in humans. However, AAV's limited cargo capacity prevents its application to therapies of inherited retinal diseases due to mutations of genes over 5 kb, like Stargardt's disease (STGD) and Usher syndrome type IB (USH1B). Previous methods based on 'forced' packaging of large genes into AAV capsids may not be easily translated to the clinic due to the generation of genomes of heterogeneous size which raise safety concerns. Taking advantage of AAV's ability to concatemerize, we generated dual AAV vectors which reconstitute a large gene by either splicing (trans-splicing), homologous recombination (overlapping), or a combination of the two (hybrid). We found that dual trans-splicing and hybrid vectors transduce efficiently mouse and pig photoreceptors to levels that, albeit lower than those achieved with a single AAV, resulted in significant improvement of the retinal phenotype of mouse models of STGD and USH1B. Thus, dual AAV trans-splicing or hybrid vectors are an attractive strategy for gene therapy of retinal diseases that require delivery of large genes.


Subject(s)
Dependovirus/metabolism , Gene Transfer Techniques , Genetic Vectors/metabolism , Retina/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Disease Models, Animal , Genetic Vectors/administration & dosage , HEK293 Cells , Humans , Injections , Lipofuscin/metabolism , Macular Degeneration/congenital , Macular Degeneration/genetics , Macular Degeneration/therapy , Melanosomes/metabolism , Mice , Mice, Inbred C57BL , Myosin VIIa , Myosins/metabolism , Phenotype , Photoreceptor Cells, Vertebrate/metabolism , Retinal Pigment Epithelium/metabolism , Rhodopsin/metabolism , Stargardt Disease , Sus scrofa , Trans-Splicing/genetics , Transduction, Genetic , Usher Syndromes/genetics , Usher Syndromes/therapy
9.
Hum Mol Genet ; 20(11): 2251-62, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21421996

ABSTRACT

Given the high genetic heterogeneity of inherited retinal degenerations (IRDs), a wide applicable treatment would be desirable to halt/slow progressive photoreceptor (PR) cell loss in a mutation-independent manner. In addition to its erythropoietic activity, erythropoietin (EPO) presents neurotrophic characteristics. We have previously shown that adeno-associated viral (AAV) vector-mediated systemic EPO delivery protects from PR degeneration. However, this is associated with an undesired hematocrit increase that could contribute to PR protection. Non-erythropoietic EPO derivatives (EPO-D) are available which allow us to dissect erythropoiesis's role in PR preservation and may be more versatile and safe than EPO as anti-apoptotic agents. We delivered in animal models of light-induced or genetic retinal degeneration either intramuscularly or subretinally AAV vectors encoding EPO or one of the three selected EPO-D: the mutant S100E, the helix A- and B-derived EPO-mimetic peptides. We observed that (i) systemic expression of S100E induces a significantly lower hematocrit increase than EPO and provides similar protection from PR degeneration, and (ii) intraocular expression of EPO-D protects PR from degeneration in the absence of significant hematocrit increase. On the basis of this, we conclude that erythropoiesis is not required for EPO-mediated PR protection. However, the lower efficacy observed when EPO or S100E is expressed intraocularly rather than systemically suggests that hormone systemic effects contribute to PR protection. Unlike S100E, EPO-mimetic peptides preserve PR only when given locally, suggesting that different EPO-D have a different potency or mode of action. In conclusion, our data show that subretinal delivery of AAV vectors encoding EPO-D protects from light-induced and genetic PR degeneration.


Subject(s)
Erythropoietin/pharmacology , Light/adverse effects , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/therapy , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Dependovirus , Erythropoiesis , Erythropoietin/genetics , Gene Transfer Techniques , Genetic Vectors , Hematocrit , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Models, Animal , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Peripherins , Rats , Rats, Inbred Lew , Retinal Degeneration/genetics
10.
Invest Ophthalmol Vis Sci ; 52(8): 5713-9, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21273543

ABSTRACT

PURPOSE: Mutations in the PDE6B gene cause recessive, severe retinitis pigmentosa (RP). PDE6B encodes the ß subunit of the rod-specific phosphodiesterase (ßPDE), which, when absent, results in toxic levels of intracellular Ca(2+) and photoreceptor cell death. Ca(2+) blockers, such as nilvadipine, as well as light restriction, slow photoreceptor degeneration in animal models of ßPDE deficiencies. The goal of the study was to evaluate the efficacy of AAV2/5- or AAV2/8-mediated gene replacement in combination with nilvadipine and/or with light restriction in the rd10 mouse bearing homozygous pde6b mutations. METHODS: AAV vectors encoding either ßPDE or EGFP were subretinally administered at postnatal day (P)2. Nilvadipine was administered from P7 to P28. For light restriction, pregnant rd10 mice were kept in a dark environment until their pups were 28 days old. All functional and histologic analyses were performed at P35. RESULTS: Significant morphologic photoreceptor protection was observed after subretinal administration of AAV vectors encoding EGFP. This protection further increased after administration of AAV2/8 or -2/5 encoding for ßPDE and was not associated with significant functional improvement. Photoreceptor protection was higher after AAV2/8- than after AAV2/5-mediated delivery and was not significantly augmented by additional drug therapy and/or light restriction. The protective effect was lost after P35. CONCLUSIONS: In conclusion, more efficient gene transfer tools than those used in this study, as well as a better understanding of the disease pathogenesis, should be explored to increase the effect of gene replacement and to design gene-based strategies that block the apoptotic pathways activated by ßPDE deficiency.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Genetic Therapy/methods , Nifedipine/analogs & derivatives , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Animals , COS Cells , Calcium Channel Blockers/pharmacology , Chlorocebus aethiops , Combined Modality Therapy , Darkness , Dependovirus/genetics , Disease Models, Animal , Female , Genetic Vectors/genetics , Homozygote , Intravitreal Injections , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nifedipine/pharmacology , Photoreceptor Cells, Vertebrate/pathology , Pregnancy , Retinitis Pigmentosa/drug therapy
11.
J Virol ; 81(20): 11372-80, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17699581

ABSTRACT

Severe inherited retinal diseases, such as retinitis pigmentosa and Leber congenital amaurosis, are caused by mutations in genes preferentially expressed in photoreceptors. While adeno-associated virus (AAV)-mediated gene transfer can correct retinal pigment epithelium (RPE) defects in animal models, approaches for the correction of photoreceptor-specific diseases are less efficient. We evaluated the ability of novel AAV serotypes (AAV2/7, AAV2/8, AAV2/9, AAV2rh.43, AAV2rh.64R1, and AAV2hu.29R) in combination with constitutive or photoreceptor-specific promoters to improve photoreceptor transduction, a limiting step in photoreceptor rescue. Based on a qualitative analysis, all AAV serotypes tested efficiently transduce the RPE as well as rod and cone photoreceptors after subretinal administration in mice. Interestingly, AAV2/9 efficiently transduces Müller cells. To compare photoreceptor transduction from different AAVs and promoters in both a qualitative and quantitative manner, we designed a strategy based on the use of a bicistronic construct expressing both enhanced green fluorescent protein and luciferase. We found that AAV2/8 and AAV2/7 mediate six- to eightfold higher levels of in vivo photoreceptor transduction than AAV2/5, considered so far the most efficient AAV serotype for photoreceptor targeting. In addition, following subretinal administration of AAV, the rhodopsin promoter allows significantly higher levels of photoreceptor expression than the other ubiquitous or photoreceptor-specific promoters tested. Finally, we show that AAV2/7, AAV2/8, and AAV2/9 outperform AAV2/5 following ex vivo transduction of retinal progenitor cells differentiated into photoreceptors. We conclude that AAV2/7 or AAV2/8 and the rhodopsin promoter provide the highest levels of photoreceptor transduction both in and ex vivo and that this may overcome the limitation to therapeutic success observed so far in models of inherited severe photoreceptor diseases.


Subject(s)
Dependovirus/genetics , Photoreceptor Cells/metabolism , Transduction, Genetic/methods , Animals , Genetic Vectors , Mice , Promoter Regions, Genetic , Retina/cytology , Rhodopsin/genetics , Serotyping , Transduction, Genetic/standards
SELECTION OF CITATIONS
SEARCH DETAIL