Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
Add more filters

Publication year range
1.
Mol Ther ; 31(7): 2042-2055, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37016576

ABSTRACT

We reported previously that ß-site amyloid precursor protein cleaving enzyme (BACE1) is strongly expressed in the normal retina and that BACE1-/- mice develop pathological phenotypes associated with age-related macular degeneration (AMD). BACE1 expression is increased within the neural retina and retinal pigment epithelium (RPE) in AMD donor eyes suggesting that increased BACE1 is compensatory. We observed that AAV-mediated BACE1 overexpression in the RPE was maintained up to 6 months after AAV1-BACE1 administration. No significant changes in normal mouse visual function or retinal morphology were observed with low-dose vector while the high-dose vector demonstrated some early pathology which regressed with time. No increase in ß-amyloid was observed. BACE1 overexpression in the RPE of the superoxide dismutase 2 knockdown (SOD2 KD) mouse, which exhibits an AMD-like phenotype, prevented loss of retinal function and retinal pathology, and this was sustained out to 6 months. Furthermore, BACE1 overexpression was able to inhibit oxidative stress, microglial changes, and loss of RPE tight junction integrity (all features of AMD) in SOD2 KD mice. In conclusion, BACE1 plays a key role in retina/RPE homeostasis, and BACE1 overexpression offers a novel therapeutic target in the treatment of AMD.


Subject(s)
Amyloid Precursor Protein Secretases , Macular Degeneration , Animals , Mice , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/genetics , Macular Degeneration/genetics , Macular Degeneration/prevention & control , Retina/metabolism , Retinal Pigment Epithelium/metabolism
2.
Mol Ther ; 31(12): 3441-3456, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37814449

ABSTRACT

Adeno-associated virus (AAV) continues to be the gold standard vector for therapeutic gene delivery and has proven especially useful for treating ocular disease. Intravitreal injection (IVtI) is a promising delivery route because it increases accessibility of gene therapies to larger patient populations. However, data from clinical and non-human primate (NHP) studies utilizing currently available capsids indicate that anatomical barriers to AAV and pre-existing neutralizing antibodies can restrict gene expression to levels that are "sub-therapeutic" in a substantial proportion of patients. Here, we performed a combination of directed evolution in NHPs of an AAV2-based capsid library with simultaneous mutations across six surface-exposed variable regions and rational design to identify novel capsid variants with improved retinal transduction following IVtI. Following two rounds of screening in NHP, enriched variants were characterized in intravitreally injected mice and NHPs and shown to have increased transduction relative to AAV2. Lead capsid variant, P2-V1, demonstrated an increased ability to evade neutralizing antibodies in human vitreous samples relative to AAV2 and AAV2.7m8. Taken together, this study further contributed to our understanding of the selective pressures associated with retinal transduction via the vitreous and identified promising novel AAV capsid variants for clinical consideration.


Subject(s)
Antibodies, Neutralizing , Capsid , Humans , Mice , Animals , Dependovirus , Intravitreal Injections , Transduction, Genetic , Primates/genetics , Capsid Proteins/genetics , Capsid Proteins/metabolism , Genetic Vectors/genetics
3.
Adv Exp Med Biol ; 1415: 125-130, 2023.
Article in English | MEDLINE | ID: mdl-37440024

ABSTRACT

Myosin VIIA (MYO7A)-associated Usher syndrome type 1B (USH1B) is a severe disorder that impacts the auditory, vestibular, and visual systems of affected patients. Due to the large size (~7.5 kb) of the MYO7A coding sequence, we have designed a dual adeno-associated virus (AAV) vector-based approach for the treatment of USH1B-related vision loss. Due to the added complexity of dual-AAV gene therapy, careful attention must be paid to the protein products expressed following vector recombination. In order to improve the sensitivity and quantifiability of our immunoassays, we adapted our traditional western blot protocol for use with the Jess™ Simple Western System. Following several rounds of testing, we optimized our protocol for the detection of MYO7A in two of our most frequently used sample types, mouse eyes, and infected HEK293 cell lysates.


Subject(s)
Myosins , Usher Syndromes , Mice , Animals , Humans , Myosins/genetics , Myosins/metabolism , HEK293 Cells , Usher Syndromes/genetics , Usher Syndromes/therapy , Myosin VIIa/genetics , Mutation
4.
J Virol ; 95(10)2021 04 26.
Article in English | MEDLINE | ID: mdl-33658343

ABSTRACT

Adeno-associated viruses (AAVs) have recently emerged as the leading vector for retinal gene therapy. However, AAV vectors which are capable of achieving clinically relevant levels of transgene expression and widespread retinal transduction are still an unmet need. Using rationally designed AAV2-based capsid variants, we investigate the role of capsid hydrophilicity and hydrophobicity as it relates to retinal transduction. We show that hydrophilic, single amino acid (aa) mutations (V387R, W502H, E530K, L583R) in AAV2 negatively impact retinal transduction when heparan sulfate proteoglycan (HSPG) binding remains intact. Conversely, addition of hydrophobic point mutations to an HSPG binding deficient capsid (AAV2ΔHS) lead to increased retinal transduction in both mouse and macaque. Our top performing vector, AAV2(4pMut)ΔHS, achieved robust rod and cone photoreceptor (PR) transduction in macaque, especially in the fovea, and demonstrates the ability to spread laterally beyond the borders of the subretinal injection (SRI) bleb. This study both evaluates biophysical properties of AAV capsids that influence retinal transduction, and assesses the transduction and tropism of a novel capsid variant in a clinically relevant animal model.ImportanceRationally guided engineering of AAV capsids aims to create new generations of vectors with enhanced potential for human gene therapy. By applying rational design principles to AAV2-based capsids, we evaluated the influence of hydrophilic and hydrophobic amino acid (aa) mutations on retinal transduction as it relates to vector administration route. Through this approach we identified a largely deleterious relationship between hydrophilic aa mutations and canonical HSPG binding by AAV2-based capsids. Conversely, the inclusion of hydrophobic aa substitutions on a HSPG binding deficient capsid (AAV2ΔHS), generated a vector capable of robust rod and cone photoreceptor (PR) transduction. This vector AAV2(4pMut)ΔHS also demonstrates a remarkable ability to spread laterally beyond the initial subretinal injection (SRI) bleb, making it an ideal candidate for the treatment of retinal diseases which require a large area of transduction.

5.
Mol Ther ; 29(8): 2456-2468, 2021 08 04.
Article in English | MEDLINE | ID: mdl-33781914

ABSTRACT

The inherited childhood blindness caused by mutations in NPHP5, a form of Leber congenital amaurosis, results in abnormal development, dysfunction, and degeneration of photoreceptors. A naturally occurring NPHP5 mutation in dogs leads to a phenotype that very nearly duplicates the human retinopathy in terms of the photoreceptors involved, spatial distribution of degeneration, and the natural history of vision loss. We show that adeno-associated virus (AAV)-mediated NPHP5 gene augmentation of mutant canine retinas at the time of active degeneration and peak cell death stably restores photoreceptor structure, function, and vision with either the canine or human NPHP5 transgenes. Mutant cone photoreceptors, which failed to form outer segments during development, reform this structure after treatment. Degenerating rod photoreceptor outer segments are stabilized and develop normal structure. This process begins within 8 weeks after treatment and remains stable throughout the 6-month posttreatment period. In both photoreceptor cell classes mislocalization of rod and cone opsins is minimized or reversed. Retinal function and functional vision are restored. Efficacy of gene therapy in this large animal ciliopathy model of Leber congenital amaurosis provides a path for translation to human treatment.


Subject(s)
Calmodulin-Binding Proteins/administration & dosage , Dependovirus/genetics , Leber Congenital Amaurosis/therapy , Retinal Cone Photoreceptor Cells/pathology , Animals , Calmodulin-Binding Proteins/pharmacology , Disease Models, Animal , Dogs , Electroretinography , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Leber Congenital Amaurosis/genetics , Treatment Outcome
6.
Mol Ther ; 29(2): 464-488, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33309881

ABSTRACT

Hereditary diseases are caused by mutations in genes, and more than 7,000 rare diseases affect over 30 million Americans. For more than 30 years, hundreds of researchers have maintained that genetic modifications would provide effective treatments for many inherited human diseases, offering durable and possibly curative clinical benefit with a single treatment. This review is limited to gene therapy using adeno-associated virus (AAV) because the gene delivered by this vector does not integrate into the patient genome and has a low immunogenicity. There are now five treatments approved for commercialization and currently available, i.e., Luxturna, Zolgensma, the two chimeric antigen receptor T cell (CAR-T) therapies (Yescarta and Kymriah), and Strimvelis (the gammaretrovirus approved for adenosine deaminase-severe combined immunodeficiency [ADA-SCID] in Europe). Dozens of other treatments are under clinical trials. The review article presents a broad overview of the field of therapy by in vivo gene transfer. We review gene therapy for neuromuscular disorders (spinal muscular atrophy [SMA]; Duchenne muscular dystrophy [DMD]; X-linked myotubular myopathy [XLMTM]; and diseases of the central nervous system, including Alzheimer's disease, Parkinson's disease, Canavan disease, aromatic l-amino acid decarboxylase [AADC] deficiency, and giant axonal neuropathy), ocular disorders (Leber congenital amaurosis, age-related macular degeneration [AMD], choroideremia, achromatopsia, retinitis pigmentosa, and X-linked retinoschisis), the bleeding disorder hemophilia, and lysosomal storage disorders.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Genetic Vectors/genetics , Animals , Clinical Studies as Topic , Combined Modality Therapy , Gene Expression , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Therapy/trends , Genetic Vectors/administration & dosage , Humans , Organ Specificity , Treatment Outcome
7.
Gene Ther ; 28(7-8): 447-455, 2021 08.
Article in English | MEDLINE | ID: mdl-33244179

ABSTRACT

Mucopolysaccharidosis type IIIB (MPS IIIB) is an autosomal recessive lysosomal disease caused by defective production of the enzyme α-N-acetylglucosaminidase. It is characterized by severe and complex central nervous system degeneration. Effective therapies will likely target early onset disease and overcome the blood-brain barrier. Modifications of adeno-associated viral (AAV) vector capsids that enhance transduction efficiency have been described in the retina. Herein, we describe for the first time, a transduction assessment of two intracranially administered adeno-associated virus serotype 8 variants, in which specific surface-exposed tyrosine (Y) and threonine (T) residues were substituted with phenylalanine (F) and valine (V) residues, respectively. A double-mutant (Y444 + 733F) and a triple-mutant (Y444 + 733F + T494V) AAV8 were evaluated for their efficacy for the potential treatment of MPS IIIB in a neonatal setting. We evaluated biodistribution and transduction profiles of both variants compared to the unmodified parental AAV8, and assessed whether the method of vector administration would modulate their utility. Vectors were administered through four intracranial routes: six sites (IC6), thalamic (T), intracerebroventricular, and ventral tegmental area into neonatal mice. Overall, we conclude that the IC6 method resulted in the widest biodistribution within the brain. Noteworthy, we demonstrate that GFP intensity was significantly more robust with AAV8 (double Y-F + T-V) compared to AAV8 (double Y-F). This provides proof of concept for the enhanced utility of IC6 administration of the capsid modified AAV8 (double Y-F + T-V) as a valid therapeutic approach for the treatment of MPS IIIB, with further implications for other monogenic diseases.


Subject(s)
Capsid , Mucopolysaccharidosis III , Animals , Brain , Dependovirus/genetics , Genetic Vectors/genetics , Mice , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/therapy , Tissue Distribution , Transduction, Genetic
8.
Mol Ther ; 28(6): 1464-1478, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32304666

ABSTRACT

The majority of inherited retinal diseases (IRDs) are caused by mutations in genes expressed in photoreceptors (PRs). The ideal vector to address these conditions is one that transduces PRs in large areas of retina with the smallest volume/lowest titer possible, and efficiently transduces foveal cones, the cells responsible for acute, daylight vision that are often the only remaining area of functional retina in IRDs. The purpose of our study was to evaluate the retinal tropism and potency of a novel capsid, AAV44.9, and rationally designed derivatives thereof. We found that AAV44.9 and AAV44.9(E531D) transduced retinas of subretinally injected (SRI) mice with higher efficiency than did benchmark AAV5- and AAV8-based vectors. In macaques, highly efficient cone and rod transduction was observed following submacular and peripheral SRI. AAV44.9- and AAV44.9(E531D)-mediated GFP fluorescence extended laterally well beyond SRI bleb margins. Notably, extrafoveal injection (i.e., fovea not detached during surgery) led to transduction of up to 98% of foveal cones. AAV44.9(E531D) efficiently transduced parafoveal and perifoveal cones, whereas AAV44.9 did not. AAV44.9(E531D) was also capable of restoring retinal function to a mouse model of IRD. These novel capsids will be useful for addressing IRDs that would benefit from an expansive treatment area.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/genetics , Retina/metabolism , Transduction, Genetic , Animals , Dependovirus/classification , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression , Genes, Reporter , Genetic Engineering , Genetic Vectors/administration & dosage , Injections, Intraocular , Macaca fascicularis , Mice , Microscopy, Confocal , Ophthalmoscopy , Promoter Regions, Genetic , Retinal Cone Photoreceptor Cells/metabolism , Retinal Diseases/genetics , Retinal Diseases/pathology , Retinal Diseases/therapy , Retinal Rod Photoreceptor Cells/metabolism , Transgenes
9.
Mol Ther ; 26(10): 2407-2417, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30078764

ABSTRACT

Retinal degenerations are a large cluster of diseases characterized by the irreversible loss of light-sensitive photoreceptors that impairs the vision of 9.1 million people in the US. An attractive treatment option is to use gene therapy to deliver broad-spectrum neuroprotective factors. However, this approach has had limited clinical translation because of the inability to control transgene expression. To address this problem, we generated an adeno-associated virus vector named RPF2 that was engineered to express domains of leukemia inhibitory factor fused to the destabilization domain of bacterial dihydrofolate reductase. Fusion proteins containing the destabilization domain are degraded in mammalian cells but can be stabilized with the binding of the drug trimethoprim. Our data show that expression levels of RPF2 are tightly regulated by the dose of trimethoprim and can be reversed by trimethoprim withdrawal. We further show that stabilized RPF2 can protect photoreceptors and prevent blindness in treated mice.


Subject(s)
Genetic Therapy , Leukemia Inhibitory Factor/genetics , Retinal Degeneration/therapy , Animals , Dependovirus/genetics , Gene Expression Regulation/drug effects , Humans , Leukemia Inhibitory Factor/administration & dosage , Mice , Neuroprotection/genetics , Photoreceptor Cells/drug effects , Photoreceptor Cells/pathology , Retina/drug effects , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Tetrahydrofolate Dehydrogenase/genetics , Transgenes/drug effects , Trimethoprim/administration & dosage
10.
Mol Ther ; 26(10): 2379-2396, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30217731

ABSTRACT

Dysregulation of the complement system is implicated in neurodegeneration, including human and animal glaucoma. Optic nerve and retinal damage in glaucoma is preceded by local complement upregulation and activation, but whether targeting this early innate immune response could have therapeutic benefit remains undefined. Because complement signals through three pathways that intersect at complement C3 activation, here we targeted this step to restore complement balance in the glaucomatous retina and to determine its contribution to degeneration onset and/or progression. To achieve this, we combined adeno-associated virus retinal gene therapy with the targeted C3 inhibitor CR2-Crry. We show that intravitreal injection of AAV2.CR2-Crry produced sustained Crry overexpression in the retina and reduced deposition of the activation product complement C3d on retinal ganglion cells and the inner retina of DBA/2J mice. This resulted in neuroprotection of retinal ganglion cell axons and somata despite continued intraocular pressure elevation, suggesting a direct restriction of neurodegeneration onset and progression and significant delay to terminal disease stages. Our study uncovers a damaging effect of complement C3 or downstream complement activation in glaucoma, and it establishes AAV2.CR2-Crry as a viable therapeutic strategy to target pathogenic C3-mediated complement activation in the glaucomatous retina.


Subject(s)
Complement C3/genetics , Glaucoma/therapy , Nerve Degeneration/therapy , Recombinant Fusion Proteins/genetics , Animals , Complement C3/antagonists & inhibitors , Dependovirus/genetics , Disease Models, Animal , Disease Progression , Gene Expression Regulation/drug effects , Genetic Therapy , Glaucoma/genetics , Glaucoma/pathology , Humans , Intraocular Pressure/drug effects , Intravitreal Injections , Mice , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Recombinant Fusion Proteins/administration & dosage , Retina/drug effects , Retina/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology
12.
Mol Ther ; 25(8): 1866-1880, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28566226

ABSTRACT

X-linked retinitis pigmentosa (XLRP) caused by mutations in the RPGR gene is an early onset and severe cause of blindness. Successful proof-of-concept studies in a canine model have recently shown that development of a corrective gene therapy for RPGR-XLRP may now be an attainable goal. In preparation for a future clinical trial, we have here optimized the therapeutic AAV vector construct by showing that GRK1 (rather than IRBP) is a more efficient promoter for targeting gene expression to both rods and cones in non-human primates. Two transgenes were used in RPGR mutant (XLPRA2) dogs under the control of the GRK1 promoter. First was the previously developed stabilized human RPGR (hRPGRstb). Second was a new full-length stabilized and codon-optimized human RPGR (hRPGRco). Long-term (>2 years) studies with an AAV2/5 vector carrying hRPGRstb under control of the GRK1 promoter showed rescue of rods and cones from degeneration and retention of vision. Shorter term (3 months) studies demonstrated comparable preservation of photoreceptors in canine eyes treated with an AAV2/5 vector carrying either transgene under the control of the GRK1 promoter. These results provide the critical molecular components (GRK1 promoter, hRPGRco transgene) to now construct a therapeutic viral vector optimized for RPGR-XLRP patients.


Subject(s)
Carrier Proteins/genetics , Eye Proteins/genetics , Genes, X-Linked , Genetic Therapy , Mutation , Retina/metabolism , Retinitis Pigmentosa/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Dogs , G-Protein-Coupled Receptor Kinase 1/genetics , Gene Expression , Gene Order , Genes, Reporter , Genetic Vectors/genetics , Humans , Phenotype , Photoreceptor Cells, Vertebrate/metabolism , Primates , Promoter Regions, Genetic , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/therapy , Transduction, Genetic , Transgenes , Vision Tests
13.
Adv Exp Med Biol ; 1074: 125-131, 2018.
Article in English | MEDLINE | ID: mdl-29721936

ABSTRACT

Leber congenital amaurosis (LCA) is a devastating pediatric retinal degenerative disease, accounting for 20% of blindness in children attending schools for the blind. Mutations in the RPE65 gene, which encodes the retinal pigment epithelium-specific isomerohydrolase RPE65, account for 16% of all LCA cases. Recent findings have linked cone photoreceptor viability to thyroid hormone (TH) signaling. TH signaling regulates cell proliferation, differentiation, and metabolism. At the cellular level, TH action is regulated by the two iodothyronine deiodinases, DIO2 and DIO3. DIO2 converts the prohormone thyroxine (T4) to the bioactive hormone triiodothyronine (T3), and DIO3 inactivates T3 and T4. The present work investigates the effects of overexpression of DIO3 to suppress TH signaling and thereby modulate cone death/survival. Subretinal delivery of AAV5-IRBP/GNAT2-hDIO3 induced robust expression of DIO3 in the mouse retina and significantly reduced the number of TUNEL-positive cells in the cone-dominant LCA model Rpe65 -/- /Nrl -/- mice. Our work shows that suppressing TH signaling by overexpression of DIO3 preserves cones, supporting that suppressing TH signaling locally in the retina may represent a treatment strategy for LCA management.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/therapeutic use , Iodide Peroxidase/therapeutic use , Leber Congenital Amaurosis/therapy , Retinal Cone Photoreceptor Cells/enzymology , cis-trans-Isomerases/deficiency , Animals , Apoptosis , Basic-Leucine Zipper Transcription Factors/deficiency , Eye Proteins/genetics , Gene Expression , Genes, Synthetic , Genetic Vectors/administration & dosage , Heterotrimeric GTP-Binding Proteins/genetics , Injections, Intraocular , Iodide Peroxidase/biosynthesis , Iodide Peroxidase/genetics , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/pathology , Mice , Mice, Knockout , Mutation , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Retinal Cone Photoreceptor Cells/pathology , Retinol-Binding Proteins/genetics , Thyroid Hormones/metabolism , Transduction, Genetic
14.
Proc Natl Acad Sci U S A ; 112(42): E5689-98, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26438859

ABSTRACT

Considerable evidence supports mutations in mitochondrial genes as the cause of maternally inherited diseases affecting tissues that rely primarily on oxidative energy metabolism, usually the nervous system, the heart, and skeletal muscles. Mitochondrial diseases are diverse, and animal models currently are limited. Here we introduced a mutant human mitochondrial gene responsible for Leber hereditary optic neuropathy (LHON) into the mouse germ line using fluorescence imaging for tissue-specific enrichment in the target retinal ganglion cells. A mitochondria-targeted adeno-associated virus (MTS-AAV) containing the mutant human NADH ubiquinone oxidoreductase subunit 4 (ND4) gene followed by mitochondrial-encoded mCherry was microinjected into zygotes. Female founders with mCherry fluorescence on ophthalmoscopy were backcrossed with normal males for eight generations. Mutant human ND4 DNA was 20% of mouse ND4 and did not integrate into the host genome. Translated human ND4 protein assembled into host respiratory complexes, decreasing respiratory chain function and increasing oxidative stress. Swelling of the optic nerve head was followed by progressive demise of ganglion cells and their axons, the hallmarks of human LHON. Early visual loss that began at 3 mo and progressed to blindness 8 mo after birth was reversed by intraocular injection of MTS-AAV expressing wild-type human ND4. The technology of introducing human mitochondrial genes into the mouse germ line has never been described, to our knowledge, and has implications not only for creating animal models recapitulating the counterpart human disorder but more importantly for reversing the adverse effects of the mutant gene using gene therapy to deliver the wild-type allele.


Subject(s)
DNA, Mitochondrial/genetics , Gene Transfer Techniques , Germ Cells , Mutation , Zygote , Animals , Axons , Brain/pathology , Electron Transport , Humans , Mice , Mice, Transgenic , NADH Dehydrogenase/genetics , Oxidative Stress , Retinal Degeneration/genetics
15.
Proc Natl Acad Sci U S A ; 112(43): E5844-53, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26460017

ABSTRACT

Inherited retinal degenerations cause progressive loss of photoreceptor neurons with eventual blindness. Corrective or neuroprotective gene therapies under development could be delivered at a predegeneration stage to prevent the onset of disease, as well as at intermediate-degeneration stages to slow the rate of progression. Most preclinical gene therapy successes to date have been as predegeneration interventions. In many animal models, as well as in human studies, to date, retinal gene therapy administered well after the onset of degeneration was not able to modify the rate of progression even when successfully reversing dysfunction. We evaluated consequences of gene therapy delivered at intermediate stages of disease in a canine model of X-linked retinitis pigmentosa (XLRP) caused by a mutation in the Retinitis Pigmentosa GTPase Regulator (RPGR) gene. Spatiotemporal natural history of disease was defined and therapeutic dose selected based on predegeneration results. Then interventions were timed at earlier and later phases of intermediate-stage disease, and photoreceptor degeneration monitored with noninvasive imaging, electrophysiological function, and visual behavior for more than 2 y. All parameters showed substantial and significant arrest of the progressive time course of disease with treatment, which resulted in long-term improved retinal function and visual behavior compared with control eyes. Histology confirmed that the human RPGR transgene was stably expressed in photoreceptors and associated with improved structural preservation of rods, cones, and ON bipolar cells together with correction of opsin mislocalization. These findings in a clinically relevant large animal model demonstrate the long-term efficacy of RPGR gene augmentation and substantially broaden the therapeutic window for intervention in patients with RPGR-XLRP.


Subject(s)
Genetic Therapy , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Retinal Degeneration/therapy , Vision, Ocular , Animals , Disease Models, Animal , Dogs , Retinal Degeneration/physiopathology
16.
Hum Mol Genet ; 24(3): 670-84, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25274777

ABSTRACT

Defects in aryl hydrocarbon receptor interacting protein-like1 (AIPL1) are associated with blinding diseases with a wide range of severity in humans. We examined the mechanism behind autosomal dominant cone-rod dystrophy (adCORD) caused by 12 base pair (bp) deletion at proline 351 of hAIPL1 (P351Δ12) mutation in the primate-specific region of human AIPL1. Mutant P351Δ12 human isoform, aryl hydrocarbon receptor interacting protein-like 1 (hAIPL1) mice demonstrated a CORD phenotype with early defects in cone-mediated vision and subsequent photoreceptor degeneration. A dominant CORD phenotype was observed in double transgenic animals expressing both mutant P351Δ12 and normal hAIPL1, but not with co-expression of P351Δ12 hAIPL1 and the mouse isoform, aryl hydrocarbon receptor interacting protein-like 1 (mAipl1). Despite a dominant effect of the mutation, we successfully rescued cone-mediated vision in P351Δ12 hAIPL1 mice following high over-expression of WT hAIPL1 by adeno-associated virus-mediated gene delivery, which was stable up to 6 months after treatment. Our transgenic P351Δ12 hAIPL1 mouse offers a novel model of AIPL1-CORD, with distinct defects from both the Aipl1-null mouse mimicking LCA and the Aipl1-hypomorphic mice mimicking a slow progressing RP.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Retinitis Pigmentosa/therapy , Animals , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Female , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Mice , Mice, Transgenic , Retinal Cone Photoreceptor Cells/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Sequence Deletion
17.
Hum Mol Genet ; 24(21): 6229-39, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26310623

ABSTRACT

Adeno-associated virus (AAV) effectively targets therapeutic genes to photoreceptors, pigment epithelia, Müller glia and ganglion cells of the retina. To date, no one has shown the ability to correct, with gene replacement, an inherent defect in bipolar cells (BCs), the excitatory interneurons of the retina. Targeting BCs with gene replacement has been difficult primarily due to the relative inaccessibility of BCs to standard AAV vectors. This approach would be useful for restoration of vision in patients with complete congenital stationary night blindness (CSNB1), where signaling through the ON BCs is eliminated due to mutations in their G-protein-coupled cascade genes. For example, the majority of CSNB1 patients carry a mutation in nyctalopin (NYX), which encodes a protein essential for proper localization of the TRPM1 cation channel required for ON BC light-evoked depolarization. As a group, CSNB1 patients have a normal electroretinogram (ERG) a-wave, indicative of photoreceptor function, but lack a b-wave due to defects in ON BC signaling. Despite retinal dysfunction, the retinas of CSNB1 patients do not degenerate. The Nyx(nob) mouse model of CSNB1 faithfully mimics this phenotype. Here, we show that intravitreally injected, rationally designed AAV2(quadY-F+T-V) containing a novel 'Ple155' promoter drives either GFP or YFP_Nyx in postnatal Nyx(nob) mice. In treated Nyx(nob) retina, robust and targeted Nyx transgene expression in ON BCs partially restored the ERG b-wave and, at the cellular level, signaling in ON BCs. Our results support the potential for gene delivery to BCs and gene replacement therapy in human CSNB1.


Subject(s)
Dependovirus/genetics , Eye Diseases, Hereditary/genetics , Genetic Diseases, X-Linked/genetics , Genetic Vectors , Myopia/genetics , Night Blindness/genetics , Proteoglycans/genetics , Retinal Bipolar Cells/metabolism , Animals , Disease Models, Animal , Eye Diseases, Hereditary/metabolism , Genetic Diseases, X-Linked/metabolism , Humans , Intravitreal Injections , Mice , Mice, Inbred C57BL , Mutation , Myopia/metabolism , Night Blindness/metabolism , Promoter Regions, Genetic , Retina/metabolism , Transfection , Transgenes , Vision, Ocular
18.
J Virol ; 90(8): 4215-4231, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26865709

ABSTRACT

UNLABELLED: Adeno-associated viruses (AAVs) currently are being developed to efficiently transduce the retina following noninvasive, intravitreal (Ivt) injection. However, a major barrier encountered by intravitreally delivered AAVs is the inner limiting membrane (ILM), a basement membrane rich in heparan sulfate (HS) proteoglycan. The goal of this study was to determine the impact of HS binding on retinal transduction by Ivt-delivered AAVs. The heparin affinities of AAV2-based tyrosine-to-phenylalanine (Y-F) and threonine-to-valine (T-V) capsid mutants, designed to avoid proteasomal degradation during cellular trafficking, were established. In addition, the impact of grafting HS binding residues onto AAV1, AAV5, and AAV8(Y733F) as well as ablation of HS binding by AAV2-based vectors on retinal transduction was investigated. Finally, the potential relationship between thermal stability of AAV2-based capsids and Ivt-mediated transduction was explored. The results show that the Y-F and T-V AAV2 capsid mutants bind heparin but with slightly reduced affinity relative to that of AAV2. The grafting of HS binding increased Ivt transduction by AAV1 but not by AAV5 or AAV8(Y733F). The substitution of any canonical HS binding residues ablated Ivt-mediated transduction by AAV2-based vectors. However, these same HS variant vectors displayed efficient retinal transduction when delivered subretinally. Notably, a variant devoid of canonical HS binding residues, AAV2(4pMut)ΔHS, was remarkably efficient at transducing photoreceptors. The disparate AAV phenotypes indicate that HS binding, while critical for AAV2-based vectors, is not the sole determinant for transduction via the Ivt route. Finally, Y-F and T-V mutations alter capsid stability, with a potential relationship existing between stability and improvements in retinal transduction by Ivt injection. IMPORTANCE: AAV has emerged as the vector of choice for gene delivery to the retina, with attention focused on developing vectors that can mediate transduction following noninvasive, intravitreal injection. HS binding has been postulated to play a role in intravitreally mediated transduction of retina. Our evaluation of the HS binding of AAV2-based variants and other AAV serotype vectors and the correlation of this property with transduction points to HS affinity as a factor controlling retinal transduction following Ivt delivery. However, HS binding is not the only requirement for improved Ivt-mediated transduction. We show that AAV2-based vectors lacking heparin binding transduce retina by subretinal injection and display a remarkable ability to transduce photoreceptors, indicating that other receptors are involved in this phenotype.


Subject(s)
Dependovirus/physiology , Genetic Vectors , Heparitin Sulfate/pharmacology , Retina/metabolism , Transduction, Genetic , Animals , Capsid/metabolism , Dependovirus/drug effects , Dependovirus/genetics , Genetic Vectors/drug effects , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Humans , Injections, Intraocular , Injections, Intravenous , Liver/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Photoreceptor Cells, Vertebrate/metabolism , Reassortant Viruses/drug effects , Reassortant Viruses/genetics , Reassortant Viruses/physiology , Vitreous Body/metabolism
19.
Exp Eye Res ; 165: 175-181, 2017 12.
Article in English | MEDLINE | ID: mdl-28974356

ABSTRACT

Retinal degenerations, including age-related macular degeneration and the retinitis pigmentosa family of diseases, are among the leading causes of legal blindness in the United States. We previously found that Stanniocalcin-1 (STC-1) reduced photoreceptor loss in the S334ter-3 and Royal College of Surgeons rat models of retinal degeneration. The results were attributed in part to a reduction in oxidative stress. Herein, we tested the hypothesis that long-term delivery of STC-1 would provide therapeutic rescue in more chronic models of retinal degeneration. To achieve sustained delivery, we produced an adeno-associated virus (AAV) construct to express STC-1 (AAV-STC-1) under the control of a retinal ganglion cell targeting promoter human synapsin 1 (hSYN1). AAV-STC-1 was injected intravitreally into the P23H-1 and S334ter-4 rhodopsin transgenic rats at postnatal day 10. Tissues were collected at postnatal day 120 for confirmation of STC-1 overexpression and histologic and molecular analysis. Electroretinography (ERG) was performed in a cohort of animals at that time. Overexpression of STC-1 resulted in a significant preservation of photoreceptors as assessed by outer nuclear thickness in the P23H-1 (P < 0.05) and the S334ter-4 (P < 0.005) models compared to controls. Additionally, retinal function was significantly improved in the P23H-1 model with overexpressed STC-1 as assessed by ERG analysis (scotopic b-wave P < 0.005 and photopic b-wave P < 0.05). Microarray analysis identified common downstream gene expression changes that occurred in both models. Genes of interest based on their function were selected for validation by quantitative real-time PCR and were significantly increased in the S334ter-4 model.


Subject(s)
Dependovirus , Glycoproteins/therapeutic use , Neuroprotective Agents/therapeutic use , Retinitis Pigmentosa/drug therapy , Animals , Disease Models, Animal , Electroretinography , Glycoproteins/administration & dosage , Neuroprotective Agents/administration & dosage , Photoreceptor Cells, Vertebrate/pathology , Rats , Rats, Transgenic , Retinitis Pigmentosa/pathology
20.
FASEB J ; 30(12): 4313-4325, 2016 12.
Article in English | MEDLINE | ID: mdl-27623928

ABSTRACT

Recent studies have implicated thyroid hormone (TH) signaling in cone photoreceptor viability. Using mouse models of retinal degeneration, we found that antithyroid treatment preserves cones. This work investigates the significance of targeting intracellular TH components locally in the retina. The cellular TH level is mainly regulated by deiodinase iodothyronine (DIO)-2 and -3. DIO2 converts thyroxine (T4) to triiodothyronine (T3), which binds to the TH receptor, whereas DIO3 degrades T3 and T4. We examined cone survival after overexpression of DIO3 and inhibition of DIO2 and demonstrated the benefits of these manipulations. Subretinal delivery of AAV5-IRBP/GNAT2-DIO3, which directs expression of human DIO3 specifically in cones, increased cone density by 30-40% in a Rpe65-/- mouse model of Lebers congenital amaurosis (LCA) and in a Cpfl1 mouse with Pde6c defect model of achromatopsia, compared with their respective untreated controls. Intravitreal and topical delivery of the DIO2 inhibitor iopanoic acid also significantly improved cone survival in the LCA model mice. Moreover, the expression levels of DIO2 and Slc16a2 were significantly higher in the diseased retinas, suggesting locally elevated TH signaling. We show that targeting DIOs protects cones, and intracellular inhibition of TH components locally in the retina may represent a novel strategy for retinal degeneration management.-Yang, F., Ma, H., Belcher, J., Butler, M. R., Redmond, T. M., Boye, S. L., Hauswirth, W. W., Ding, X.-Q. Targeting iodothyronine deiodinases locally in the retina is a therapeutic strategy for retinal degeneration.


Subject(s)
Iodide Peroxidase/metabolism , Retina/metabolism , Retinal Degeneration/metabolism , Animals , Cells, Cultured , Mice, Knockout , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , Signal Transduction/physiology , Thyroid Hormones/metabolism , Triiodothyronine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL