Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Transl Vis Sci Technol ; 12(4): 20, 2023 04 03.
Article in English | MEDLINE | ID: mdl-37070938

ABSTRACT

Purpose: The expression of silent information regulator (SIRT) 1 is reduced in diabetic retinopathy (DR). Previous studies showed that alterations in SIRT1 messenger RNA (mRNA) and protein expression are implicated in progressive inflammation and formation of retinal acellular capillaries. Treatment with the SIRT1 agonist, SRT1720, improved visual response by restoration of a- and b-wave responses on electroretinogram scotopic measurements in diabetic (db/db) mice. In this study, we investigated the effects of intravitreal SIRT1 delivery on diabetic retinal pathology. Methods: Nine-month-old db/db mice received one intravitreal injection of either AAV2-SIRT1 or AAV2-GFP control virus, and after 3 months, electroretinography and optomotor responses were measured. Their eyes were then removed and analyzed by immunohistochemistry and flow cytometry. Results: SIRT1 mRNA and protein levels were increased following AAV2-SIRT1 administration compared to control virus AAV2-GFP injected mice. IBA1+ and caspase 3 expression were decreased in retinas of db/db mice injected with AAV2-SIRT1, and reductions in scotopic a- and b-waves and high spatial frequency in optokinetic response were prevented. Retinal hypoxia inducible factor 1α (HIF-1α) protein levels were reduced in the AAV2-SIRT1-injected mice compared to control-injected mice. Using flow cytometry to assess changes in intracellular HIF-1α levels, endothelial cells (CD31+) from AAV-2 SIRT1 injected mice demonstrated reduced HIF-1α expression compared to db/db mice injected with the control virus. Conclusions: Intravitreal AAV2-SIRT1 delivery increased retina SIRT1 and transduced neural and endothelial cells, thus reversing functional damage and improving overall visual function. Translational Relevance: AAV2-SIRT1 gene therapy represents a beneficial approach for the treatment of chronic retinal conditions such as DR.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Mice , Animals , Diabetic Retinopathy/genetics , Diabetic Retinopathy/therapy , Sirtuin 1/genetics , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/therapy , Endothelial Cells/metabolism , Disease Models, Animal , RNA, Messenger
2.
Transl Vis Sci Technol ; 11(11): 8, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36355386

ABSTRACT

Purpose: Ocular hypertension is a significant risk factor for vision loss in glaucoma caused by the death of retinal ganglion cells (RGCs). We investigated whether small heat shock proteins (sHsps) expressed in RGCs protect those cells against ocular hypertension in mice. Methods: AAV2 vectors encoding genes for one of the following four human sHsps: HSPB1, HSPB4, HSPB5, or HSPB6 were constructed for RGC-specific expression. Ischemia/reperfusion was induced by elevating the intraocular pressure (IOP) to 120 mm Hg for one hour, followed by a rapid return to normal IOP. Microbeads (MB) were injected into the anterior chamber of mice to induce ocular hypertension. RGC death and glial activation were assessed by immunostaining for Brn3a, RBPMS, Iba1, and glial fibrillary acid protein in retinal flat mounts. RGC axonal defects were evaluated by anterograde transport of intravitreally injected cholera toxin-B. RGC function was assessed by pattern electroretinography. Results: Among the sHsps, HspB1 offered the best protection against RGC death from ischemia/reperfusion injury in the mouse retina. Intravitreal administration of AAV2-HSPB1 either two weeks before or one week after instituting ocular hypertension resulted in significant prevention of RGC loss. The MB-injected mice showed RGC axonal transportation defects, but AAV2-HSPB1 administration significantly inhibited this defect. AAV2-HSPB1 prevented glial activation caused by ocular hypertension. More importantly, a single injection of AAV2-HSPB1 protected RGCs long-term in MB-injected eyes. Conclusions: The administration of AAV2-HSPB1 inhibited RGC death and axonal transport defects and reduced glial activation in a mouse model of ocular hypertension. Translational Relevance: Our results suggested that the intravitreal delivery of AAV2-HSPB1 could be developed as a gene therapy to prevent vision loss on a long-term basis in glaucoma patients.


Subject(s)
Glaucoma , Ocular Hypertension , Humans , Mice , Animals , Retinal Ganglion Cells/metabolism , Axonal Transport , Ocular Hypertension/genetics , Ocular Hypertension/metabolism , Glaucoma/genetics , Glaucoma/prevention & control , Intraocular Pressure , Disease Models, Animal , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism
3.
PLoS One ; 17(5): e0269261, 2022.
Article in English | MEDLINE | ID: mdl-35639753

ABSTRACT

Glaucoma is the leading cause of irreversible blindness worldwide. Therapies for glaucoma are directed toward reducing intraocular pressure (IOP), the leading risk factor and only reliable therapeutic target via topical medications or with procedural intervention including laser or surgery. Though topical therapeutics are typically first line, less than 50% of patients take drops as prescribed. Sustained release technologies that decrease IOP for extended periods of time are being examined for clinical use. We recently identified Stanniocalcin-1, a naturally occurring hormone, as an IOP-lowering agent. Here, we show that a single injection into the anterior chamber of mice with an adeno-associated viral vector containing the transgene of stanniocalcin-1 results in diffuse and sustained expression of the protein and produces IOP reduction for up to 6 months. As the treatment effect begins to wane, IOP-lowering can be rescued with a repeat injection. Aqueous humor dynamic studies revealed an increase in outflow facility as the mechanism of action. This first-in-class therapeutic approach has the potential to improve care and reduce the rates of vision loss in the 80 million people worldwide currently affected by glaucoma.


Subject(s)
Glaucoma , Ocular Hypotension , Animals , Glaucoma/drug therapy , Glaucoma/genetics , Glycoproteins , Humans , Intraocular Pressure , Mice , Tonometry, Ocular , Transgenes
4.
Invest Ophthalmol Vis Sci ; 63(2): 5, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35103748

ABSTRACT

Purpose: Based on our preview evidence that reduced nuclear content of the transcription factor Myc-associated protein X (MAX) is an early event associated with degeneration of retinal ganglion cells (RGCs), in the present study, our purpose was to test whether the overexpression of human MAX had a neuroprotective effect against RGC injury. Methods: Overexpression of either MAX or green fluorescent protein (GFP) in the retina was achieved by intravitreal injections of recombinant adenovirus-associated viruses (rAAVs). Lister Hooded rats were used in three models of RGC degeneration: (1) cultures of retinal explants for 30 hours ex vivo from the eyes of 14-day-old rats that had received intravitreal injections of rAAV2-MAX or the control vector rAAV2-GFP at birth; (2) an optic nerve crush model, in which 1-month-old rats received intravitreal injection of either rAAV2-MAX or rAAV2-GFP and, 4 weeks later, were operated on; and (3) an ocular hypertension (OHT) glaucoma model, in which 1-month-old rats received intravitreal injection of either rAAV2-MAX or rAAV2-GFP and, 4 weeks later, were subject to cauterization of the limbal plexus. Cell death was estimated by detection of pyknotic nuclei and TUNEL technique and correlated with MAX immunocontent in an ex vivo model of retinal explants. MAX expression was detected by quantitative RT-PCR. In the OHT model, survival of RGCs was quantified by retrograde labeling with DiI or immunostaining for BRN3a at 14 days after in vivo injury. Functional integrity of RGCs was analyzed through pattern electroretinography, and damage to the optic nerve was examined in semithin sections. Results: In all three models of RGC insult, gene therapy by overexpression of MAX prevented RGC death. Also, ON degeneration and electrophysiologic deficits were prevented in the OHT model. Conclusions: Our experiments offer proof of concept for a novel neuroprotective gene therapy for glaucomatous neurodegeneration based on overexpression of MAX.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Gene Expression Regulation , Genetic Therapy/methods , Glaucoma/complications , Nerve Regeneration/genetics , Neurodegenerative Diseases/therapy , Neuroprotection/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Cell Death , Disease Models, Animal , Female , Glaucoma/genetics , Glaucoma/pathology , Male , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/genetics , Rats , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
5.
Dev Biol ; 464(2): 111-123, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32562755

ABSTRACT

The transcription factors Prdm1 (Blimp1) and Vsx2 (Chx10) work downstream of Otx2 to regulate photoreceptor and bipolar cell fates in the developing retina. Mice that lack Vsx2 fail to form bipolar cells while Prdm1 mutants form excess bipolars at the direct expense of photoreceptors. Excess bipolars in Prdm1 mutants appear to derive from rods, suggesting that photoreceptor fate remains mutable for some time after cells become specified. Here we tested whether bipolar cell fate is also plastic during development. To do this, we created a system to conditionally misexpress Prdm1 at different stages of bipolar cell development. We found that Prdm1 blocks bipolar cell formation if expressed before the fate choice decision occurred. When we misexpressed Prdm1 just after the decision to become a bipolar cell was made, some cells were reprogrammed into photoreceptors. In contrast, Prdm1 misexpression in mature bipolar cells did not affect cell fate. We also provide evidence that sustained misexpression of Prdm1 was selectively toxic to photoreceptors. Our data show that bipolar fate is malleable, but only for a short temporal window following fate specification. Prdm1 and Vsx2 act by stabilizing photoreceptor and bipolar fates in developing OTX2+ cells of the retina.


Subject(s)
Cellular Reprogramming , Gene Expression Regulation, Developmental , Photoreceptor Cells, Vertebrate/metabolism , Positive Regulatory Domain I-Binding Factor 1/biosynthesis , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Mutation , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Photoreceptor Cells, Vertebrate/cytology , Positive Regulatory Domain I-Binding Factor 1/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Hum Gene Ther ; 30(11): 1361-1370, 2019 11.
Article in English | MEDLINE | ID: mdl-31418294

ABSTRACT

Autosomal recessive Stargardt disease is the most common inherited macular degeneration in humans. It is caused by mutations in the retina-specific ATP binding cassette transporter A4 (ABCA4) that is essential for the clearance of all-trans-retinal from photoreceptor cells. Loss of this function results in the accumulation of toxic bisretinoids in the outer segment disk membranes and their subsequent transfer into adjacent retinal pigment epithelium (RPE) cells. This ultimately leads to the Stargardt disease phenotype of increased retinal autofluorescence and progressive RPE and photoreceptor cell loss. Adeno-associated virus (AAV) vectors have been widely used in gene therapeutic applications, but their limited cDNA packaging capacity of ∼4.5 kb has impeded their use for transgenes exceeding this limit. AAV dual vectors were developed to overcome this size restriction. In this study, we have evaluated the in vitro expression of ABCA4 using three options: overlap, transplicing, and hybrid ABCA4 dual vector systems. The hybrid system was the most efficient of these dual vector alternatives and used to express the full-length ABCA4 in Abca4-/- mice. The full-length ABCA4 protein correctly localized to photoreceptor outer segments. Moreover, treatment of Abca4-/- mice with this ABCA4 hybrid dual vector system resulted in a reduced accumulation of the lipofuscin/N-retinylidene-N-retinylethanolamine (A2E) autofluorescence in vivo, and retinal A2E quantification supported these findings. These results show that the hybrid AAV dual vector option is both safe and therapeutic in mice, and the delivered ABCA4 transgene is functional and has a significant effect on reducing A2E accumulation in the Abca4-/- mouse model of Stargardt disease.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/therapeutic use , Dependovirus/genetics , Genes, Recessive , Genetic Vectors/metabolism , Retina/pathology , Stargardt Disease/genetics , Stargardt Disease/therapy , Animals , Disease Models, Animal , Fluorescence , Fundus Oculi , HEK293 Cells , Humans , Mice, Inbred C57BL , Retina/metabolism , Retinoids/metabolism
7.
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
8.
Front Mol Neurosci ; 11: 233, 2018.
Article in English | MEDLINE | ID: mdl-30038560

ABSTRACT

Rod and cone phosphodiesterase 6 (PDE6) are key effector enzymes of the vertebrate phototransduction pathway. Rod PDE6 consists of two catalytic subunits PDE6α and PDE6ß and two identical inhibitory PDE6γ subunits, while cone PDE6 is composed of two identical PDE6α' catalytic subunits and two identical cone-specific PDE6γ' inhibitory subunits. Despite their prominent function in regulating cGMP levels and therefore rod and cone light response properties, it is not known how each subunit contributes to the functional differences between rods and cones. In this study, we generated an rd10/cpfl1 mouse model lacking rod PDE6ß and cone PDE6α' subunits. Both rod and cone photoreceptor cells are degenerated with age and all PDE6 subunits degrade in rd10/cpfl1 mice. We expressed cone PDE6α' in both rods and cones of rd10/cpfl1 mice by adeno-associated virus (AAV)-mediated delivery driven by the ubiquitous, constitutive small chicken ß-actin promoter. We show that expression of PDE6α' rescues rod function in rd10/cpfl1 mice, and the restoration of rod light sensitivity is attained through restoration of endogenous rod PDE6γ and formation of a functional PDE6α'γ complex. However, improved photopic cone responses were achieved only after supplementation of both cone PDE6α' and PDE6γ' subunits but not by PDE6α' treatment alone. We observed a two fold increase of PDE6α' levels in the eyes injected with both PDE6α' plus PDE6γ' relative to eyes receiving PDE6α' alone. Despite the presence of both PDE6γ' and PDE6γ, the majority of PDE6α' formed functional complexes with PDE6γ', suggesting that PDE6α' has a higher association affinity for PDE6γ' than for PDE6γ. These results suggest that the presence of PDE6γ' augments cone PDE6 assembly and enhances its stability. Our finding has important implication for gene therapy of PDE6α'-associated achromatopsia.

9.
Proc Natl Acad Sci U S A ; 115(12): E2839-E2848, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29507198

ABSTRACT

Mutations in the BEST1 gene cause detachment of the retina and degeneration of photoreceptor (PR) cells due to a primary channelopathy in the neighboring retinal pigment epithelium (RPE) cells. The pathophysiology of the interaction between RPE and PR cells preceding the formation of retinal detachment remains not well-understood. Our studies of molecular pathology in the canine BEST1 disease model revealed retina-wide abnormalities at the RPE-PR interface associated with defects in the RPE microvillar ensheathment and a cone PR-associated insoluble interphotoreceptor matrix. In vivo imaging demonstrated a retina-wide RPE-PR microdetachment, which contracted with dark adaptation and expanded upon exposure to a moderate intensity of light. Subretinal BEST1 gene augmentation therapy using adeno-associated virus 2 reversed not only clinically detectable subretinal lesions but also the diffuse microdetachments. Immunohistochemical analyses showed correction of the structural alterations at the RPE-PR interface in areas with BEST1 transgene expression. Successful treatment effects were demonstrated in three different canine BEST1 genotypes with vector titers in the 0.1-to-5E11 vector genomes per mL range. Patients with biallelic BEST1 mutations exhibited large regions of retinal lamination defects, severe PR sensitivity loss, and slowing of the retinoid cycle. Human translation of canine BEST1 gene therapy success in reversal of macro- and microdetachments through restoration of cytoarchitecture at the RPE-PR interface has promise to result in improved visual function and prevent disease progression in patients affected with bestrophinopathies.


Subject(s)
Bestrophins/genetics , Eye Diseases, Hereditary/therapy , Genetic Therapy/methods , Retinal Diseases/therapy , Animals , Dog Diseases/therapy , Dogs , Eye Diseases, Hereditary/diagnostic imaging , Eye Diseases, Hereditary/pathology , Eye Diseases, Hereditary/veterinary , Female , Genetic Vectors/pharmacology , Humans , Light , Male , Mutation , Retinal Detachment/diagnostic imaging , Retinal Detachment/pathology , Retinal Detachment/therapy , Retinal Diseases/diagnostic imaging , Retinal Diseases/pathology , Retinal Diseases/veterinary , Retinal Pigment Epithelium/pathology , Tomography, Optical Coherence
10.
Mol Vis ; 24: 17-28, 2018.
Article in English | MEDLINE | ID: mdl-29386880

ABSTRACT

Purpose: Blue cone monochromacy (BCM) is an X-linked congenital vision disorder characterized by complete loss or severely reduced L- and M-cone function. Patients with BCM display poor visual acuity, severely impaired color discrimination, myopia, nystagmus, and minimally detectable cone-mediated electroretinogram. Recent studies of patients with BCM with adaptive optics scanning laser ophthalmoscopy (AOSLO) showed that they have a disrupted cone mosaic with reduced numbers of cones in the fovea that is normally dominated by L- and M-cones. The remaining cones in the fovea have significantly shortened outer segments but retain sufficient structural integrity to serve as potential gene therapy targets. In this study, we tested whether exogenously expressed human L- and M-opsins can rescue M-cone function in an M-opsin knockout (Opn1mw-/- ) mouse model for BCM. Methods: Adeno-associated virus type 5 (AAV5) vectors expressing OPN1LW, OPN1MW, or C-terminal tagged OPN1LW-Myc, or OPN1MW-HA driven by a cone-specific promoter were injected subretinally into one eye of Opn1mw-/- mice, while the contralateral eye served as the uninjected control. Expression of cone pigments was determined with western blotting and their cellular localization identified with immunohistochemistry. M-cone function was analyzed with electroretinogram (ERG). Antibodies against cone phototransduction proteins were used to study cone outer segment (OS) morphology in untreated and treated Opn1mw-/- eyes. Results: We showed that cones in the dorsal retina of the Opn1mw-/- mouse do not form outer segments, resembling cones that lack outer segments in the human BCM fovea. We further showed that AAV5-mediated expression of either human M- or L-opsin individually or combined promotes regrowth of cone outer segments and rescues M-cone function in the treated Opn1mw-/- dorsal retina. Conclusions: Exogenously expressed human opsins can regenerate cone outer segments and rescue M-cone function in Opn1mw-/- mice, thus providing a proof-of-concept gene therapy in an animal model of BCM.


Subject(s)
Color Vision Defects/therapy , Fovea Centralis/metabolism , Genetic Therapy/methods , Retinal Photoreceptor Cell Outer Segment/metabolism , Rod Opsins/genetics , Animals , Color Vision Defects/genetics , Color Vision Defects/metabolism , Color Vision Defects/pathology , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Fovea Centralis/pathology , Gene Expression , Genetic Complementation Test , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Mice , Mice, Knockout , Ophthalmoscopy , Promoter Regions, Genetic , Retinal Photoreceptor Cell Outer Segment/pathology , Rod Opsins/metabolism , Transgenes
11.
Diabetes ; 67(4): 769-781, 2018 04.
Article in English | MEDLINE | ID: mdl-29362226

ABSTRACT

Tight junctions (TJs) involve close apposition of transmembrane proteins between cells. Although TJ proteins have been studied in detail, the role of lipids is largely unknown. We addressed the role of very long-chain (VLC ≥26) ceramides in TJs using diabetes-induced loss of the blood-retinal barrier as a model. VLC fatty acids that incorporate into VLC ceramides are produced by elongase elongation of very long-chain fatty acids protein 4 (ELOVL4). ELOVL4 is significantly reduced in the diabetic retina. Overexpression of ELOVL4 significantly decreased basal permeability, inhibited vascular endothelial growth factor (VEGF)- and interleukin-1ß-induced permeability, and prevented VEGF-induced decrease in occludin expression and border staining of TJ proteins ZO-1 and claudin-5. Intravitreal delivery of AAV2-hELOVL4 reduced diabetes-induced increase in vascular permeability. Ultrastructure and lipidomic analysis revealed that ω-linked acyl-VLC ceramides colocalize with TJ complexes. Overall, normalization of retinal ELOVL4 expression could prevent blood-retinal barrier dysregulation in diabetic retinopathy through an increase in VLC ceramides and stabilization of TJs.


Subject(s)
Blood-Retinal Barrier/metabolism , Capillary Permeability/genetics , Ceramides/metabolism , Endothelial Cells/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Retinal Vessels/metabolism , Tight Junctions/metabolism , Animals , Cattle , Claudin-5/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/etiology , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Endothelial Cells/ultrastructure , Humans , Interleukin-1beta/metabolism , Mice , Occludin/metabolism , Retina/metabolism , Retinal Vessels/ultrastructure , Tight Junctions/ultrastructure , Vascular Endothelial Growth Factor A/metabolism , Zonula Occludens-1 Protein/metabolism
12.
J Clin Invest ; 128(1): 190-206, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29202463

ABSTRACT

Retinitis pigmentosa (RP) is a major cause of blindness that affects 1.5 million people worldwide. Mutations in cyclic nucleotide-gated channel ß 1 (CNGB1) cause approximately 4% of autosomal recessive RP. Gene augmentation therapy shows promise for treating inherited retinal degenerations; however, relevant animal models and biomarkers of progression in patients with RP are needed to assess therapeutic outcomes. Here, we evaluated RP patients with CNGB1 mutations for potential biomarkers of progression and compared human phenotypes with those of mouse and dog models of the disease. Additionally, we used gene augmentation therapy in a CNGß1-deficient dog model to evaluate potential translation to patients. CNGB1-deficient RP patients and mouse and dog models had a similar phenotype characterized by early loss of rod function and slow rod photoreceptor loss with a secondary decline in cone function. Advanced imaging showed promise for evaluating RP progression in human patients, and gene augmentation using adeno-associated virus vectors robustly sustained the rescue of rod function and preserved retinal structure in the dog model. Together, our results reveal an early loss of rod function in CNGB1-deficient patients and a wide window for therapeutic intervention. Moreover, the identification of potential biomarkers of outcome measures, availability of relevant animal models, and robust functional rescue from gene augmentation therapy support future work to move CNGB1-RP therapies toward clinical trials.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/deficiency , Mutation , Nerve Tissue Proteins/deficiency , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Animals , Cyclic Nucleotide-Gated Cation Channels/metabolism , Dependovirus , Disease Models, Animal , Dogs , Female , Humans , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/therapy , Transduction, Genetic
13.
Mol Vis ; 24: 834-846, 2018.
Article in English | MEDLINE | ID: mdl-30713422

ABSTRACT

Purpose: Recessive mutations in the human IQCB1/NPHP5 gene are associated with Senior-Løken syndrome (SLS), a ciliopathy presenting with nephronophthisis and Leber congenital amaurosis (LCA). Nphp5-knockout mice develop LCA without nephronophthisis. Mutant rods rapidly degenerate while mutant cones survive for months. The purpose of this study was to reinitiate cone ciliogenesis in a Nphp5 -/-; Nrl -/- mouse with viral expression of full-length NPHP5 and rescue function. Methods: Nphp5 -/- mice were mated with Nrl -/- mice to generate Nphp5-/-; Nrl-/- double-knockouts. Nphp5-/-; Nrl-/- mice and Nphp5+/-; Nrl-/- controls were phenotyped with confocal microscopy from postnatal day 10 (P10) until 6 months of age. Nphp5-/-; Nrl-/- mice and Nphp5+/-; Nrl-/- controls were injected at P15 with self-complementary adenoassociated virus 8 (Y733F) (AAV8(Y733F)) expressing GRK1-FL-cNPHP5. Expression of mutant NPHP5 was verified with confocal microscopy and electroretinography (ERG). Results: In the Nphp5 -/- and cone-only Nphp5 -/-; Nrl -/- mice, cone outer segments did not form, but mutant cones continued to express cone pigments in the inner segments without obvious signs of cone cell death. The mutant cone outer nuclear layer (ONL) and the inner segments were stable for more than 6 months in the cone-only Nphp5 -/-; Nrl -/- retinas. Viral expression of NPHP5 initiated after eye opening showed that connecting cilia and RP1-positive axonemes were formed. Furthermore, cone pigments and other cone outer segment proteins (cone transducin and cone PDE6) were present in the nascent mutant cone outer segments, and rescued mutant cones exhibited a significant photopic b-wave (30% of Nphp5 +/-; Nrl -/- controls). Conclusions: Nphp5-/-; Nrl-/- cones persistently express cone pigments in the inner segments without obvious degeneration, providing an extended duration interval for viral gene expression. Viral expression of full-length NPHP5 initiates ciliogenesis between P15 and P60, and mutant cones are, in part, functional, encouraging future retina gene replacement therapy.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Calmodulin-Binding Proteins/genetics , Eye Proteins/genetics , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/therapy , Retinal Cone Photoreceptor Cells/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Amino Acid Sequence , Animals , Axoneme/metabolism , Axoneme/ultrastructure , Basic-Leucine Zipper Transcription Factors/deficiency , Calmodulin-Binding Proteins/deficiency , Cilia/metabolism , Cilia/ultrastructure , Crosses, Genetic , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Disease Models, Animal , Eye Proteins/metabolism , Female , G-Protein-Coupled Receptor Kinase 1/genetics , G-Protein-Coupled Receptor Kinase 1/metabolism , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , Gene Expression Regulation , Genetic Therapy/methods , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/pathology , Male , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phenotype , Retinal Cone Photoreceptor Cells/pathology , Sequence Alignment , Sequence Homology, Amino Acid , Transducin/genetics , Transducin/metabolism
14.
Hum Gene Ther Clin Dev ; 28(4): 197-207, 2017 12.
Article in English | MEDLINE | ID: mdl-29020838

ABSTRACT

Achromatopsia is an inherited retinal disorder of cone photoreceptors characterized by markedly reduced visual acuity, extreme light sensitivity, and absence of color discrimination. Approximately 50% of cases are caused by mutations in the cone photoreceptor-specific cyclic nucleotide gated channel beta subunit (CNGB3) gene. Studies in CNGB3-mutant dogs showed that subretinal injection of an AAV vector expressing human CNGB3, which has 76% amino acid identity with canine CNGB3, driven by a 2.1 kb human red cone opsin promoter (PR2.1) and packaged in AAV5 capsids (AAV5-PR2.1-hCNGB3) rescued cone photoreceptor function, but at high doses was associated with an inflammatory response (focal chorioretinitis) consistent with immune-mediated toxicity. AAV vectors containing the PR2.1 promoter packaged in AAV5 capsids and expressing either the native canine CNGB3 (AAV5-PR2.1-cCNGB3) or the human CNGB3 (AAV5-PR2.1-hCNGB3) were evaluated at different dose levels in CNGB3-mutant dogs. The vector expressing canine CNGB3 achieved somewhat better rescue of cone function but unexpectedly was associated with a greater degree of retinal toxicity than the vector expressing human CNGB3. Very low-level T-cell immune responses to some AAV or CNGB3 peptides were observed in animals that received the higher vector dose. There was a more than twofold increase in serum neutralizing antibodies to AAV in one of three animals in the low-dose group and in two of three animals in the high-dose group. No serum anti-hCNGB3 antibodies were detected in any animal. The results of this study do not support the hypothesis that the focal chorioretinitis seen with high doses of AAV5-PR2.1-hCNGB3 in the initial studies was due to an immune response to human CNGB3.


Subject(s)
Color Vision Defects/genetics , Color Vision Defects/therapy , Cyclic Nucleotide-Gated Cation Channels/therapeutic use , Genetic Therapy , Animals , Chorioretinitis/genetics , Chorioretinitis/pathology , Chorioretinitis/therapy , Color Vision Defects/pathology , Cyclic Nucleotide-Gated Cation Channels/genetics , Dependovirus , Dog Diseases/genetics , Dog Diseases/pathology , Dog Diseases/therapy , Dogs , Genetic Vectors/therapeutic use , Humans , Immunity, Cellular/genetics , Opsins/genetics , Parvovirinae/genetics , Promoter Regions, Genetic/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology
15.
Mol Ther ; 25(2): 392-400, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28153090

ABSTRACT

Brain ischemia is a major cause of adult disability and death, and it represents a worldwide health problem with significant economic burden for modern society. The identification of the molecular pathways activated after brain ischemia, together with efficient technologies of gene delivery to the CNS, may lead to novel treatments based on gene therapy. Recombinant adeno-associated virus (rAAV) is an effective platform for gene transfer to the CNS. Here, we used a serotype 8 rAAV bearing the Y733F mutation (rAAV8-733) to overexpress co-chaperone E3 ligase CHIP (also known as Stub-1) in rat hippocampal neurons, both in an oxygen and glucose deprivation model in vitro and in a four-vessel occlusion model of ischemia in vivo. We show that CHIP overexpression prevented neuronal degeneration in both cases and led to a decrease of both eIF2α (serine 51) and AKT (serine 473) phosphorylation, as well as reduced amounts of ubiquitinated proteins following hypoxia or ischemia. These data add to current knowledge of ischemia-related signaling in the brain and suggest that gene therapy based on the role of CHIP in proteostasis may provide a new venue for brain ischemia treatment.


Subject(s)
Brain Ischemia/genetics , Cell Death/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Pyramidal Cells/metabolism , Transduction, Genetic , Ubiquitin-Protein Ligases/genetics , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Dependovirus/classification , Disease Models, Animal , Gene Expression , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/administration & dosage , Glucose/metabolism , Hypoxia/metabolism , Oxygen/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Pyramidal Cells/pathology , Rats , Ubiquitination
16.
Am J Pathol ; 186(9): 2486-99, 2016 09.
Article in English | MEDLINE | ID: mdl-27423695

ABSTRACT

Occludin is a transmembrane tight junction protein that contributes to diverse cellular functions, including control of barrier properties, cell migration, and proliferation. Vascular endothelial growth factor (VEGF) induces phosphorylation of occludin at S490, which is required for VEGF-induced endothelial permeability. Herein, we demonstrate that occludin S490 phosphorylation also regulates VEGF-induced retinal endothelial cell proliferation and neovascularization. Using a specific antibody, phospho-occludin was located in centrosomes in endothelial cell cultures, animal models, and human surgical samples of retinal neovessels. Occludin S490 phosphorylation was found to increase with endothelial tube formation in vitro and in vivo during retinal neovascularization after induction of VEGF expression. More important, expression of occludin mutated at S490 to Ala, completely inhibited angiogenesis in cell culture models and in vivo. Collectively, these data suggest a novel role for occludin in regulation of endothelial proliferation and angiogenesis in a phosphorylation-dependent manner. These findings may lead to methods of regulating pathological neovascularization by specifically targeting endothelial cell proliferation.


Subject(s)
Occludin/metabolism , Retinal Neovascularization/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Blood-Retinal Barrier/metabolism , Blotting, Western , Cattle , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Phosphorylation
17.
PLoS One ; 11(2): e0148874, 2016.
Article in English | MEDLINE | ID: mdl-26881841

ABSTRACT

Usher syndrome type III (USH3A) is an autosomal recessive disorder caused by mutations in clarin-1 (CLRN1) gene, leading to progressive retinal degeneration and sensorineural deafness. Efforts to develop therapies for preventing photoreceptor cell loss are hampered by the lack of a retinal phenotype in the existing USH3 mouse models and by conflicting reports regarding the endogenous retinal localization of clarin-1, a transmembrane protein of unknown function. In this study, we used an AAV-based approach to express CLRN1 in the mouse retina in order to determine the pattern of its subcellular localization in different cell types. We found that all major classes of retinal cells express AAV-delivered CLRN1 driven by the ubiquitous, constitutive small chicken ß-actin promoter, which has important implications for the design of future USH3 gene therapy studies. Within photoreceptor cells, AAV-expressed CLRN1 is mainly localized at the inner segment region and outer plexiform layer, similar to the endogenous expression of other usher proteins. Subretinal delivery using a full strength viral titer led to significant loss of retinal function as evidenced by ERG analysis, suggesting that there is a critical limit for CLRN1 expression in photoreceptor cells. Taken together, these results suggest that CLRN1 expression is potentially supported by a variety of retinal cells, and the right combination of AAV vector dose, promoter, and delivery method needs to be selected to develop safe therapies for USH3 disorder.


Subject(s)
Genetic Therapy , Membrane Proteins/biosynthesis , Retinal Degeneration/genetics , Usher Syndromes/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Gene Expression Regulation , Humans , Membrane Proteins/genetics , Mice , Retina/metabolism , Retina/pathology , Retinal Degeneration/pathology , Retinal Degeneration/therapy , Usher Syndromes/pathology , Usher Syndromes/therapy
18.
Invest Ophthalmol Vis Sci ; 56(11): 6971-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26513502

ABSTRACT

PURPOSE: The mutation S163R in complement C1q tumor necrosis factor-related protein-5 (C1QTNF5) causes an autosomal dominant disorder known as late-onset retinal degeneration (L-ORD). In this study, our goal is to evaluate the consequences of mutant S163R C1QTNF5 expression in mouse RPE following its delivery using an adeno-associated viral (AAV) vector. METHODS: We generated AAV vectors containing either human wild-type C1QTNF5 or mutant S163R C1QTNF5 driven by an RPE-specific BEST1 promoter, and delivered them subretinally into one eye of adult C57BL/6 mice. Transgene expression was detected by immunohistochemistry. Retinal function was assessed by full-field ERG. Pathological changes were further examined by digital fundus imaging and spectral-domain optical coherence tomography (SD-OCT). RESULTS: We show that the AAV-expressed mutant S163R leads to pathological effects similar to some of those found in patients with advanced L-ORD, including RPE thinning, RPE cell loss, and retinal degeneration. In addition, we provide in vivo evidence that mutant S163R C1QTNF5 can form large, transparent, spherical intracellular aggregates throughout the RPE, which are detectable by light microscopy. In contrast to AAV-expressed wild-type C1QTNF5, which is secreted apically from the RPE toward the photoreceptor cells and the outer limiting membrane, the S163R mutant is primarily routed toward the basal side of RPE, where it forms thick, extracellular deposits over time. CONCLUSIONS: Adeno-associated viral-targeted expression of mutant S163R in the RPE represents a useful approach for quickly generating animal models that mimic pathological features of L-ORD and offers the potential to understand disease mechanisms and develop therapeutic strategies.


Subject(s)
Membrane Proteins/genetics , Retinal Pigment Epithelium/pathology , Animals , Bestrophins , Blotting, Western , Eye Proteins/genetics , Fundus Oculi , Gene Expression , Ion Channels/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Mutation, Missense , Retinal Degeneration/genetics , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure , Tomography, Optical Coherence
19.
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
20.
Hum Gene Ther ; 26(9): 593-602, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26076799

ABSTRACT

Our collaborative successful gene replacement therapy using AAV vectors expressing a variant of human RPGR-ORF15 in two canine models provided therapeutic proof of concept for translation into human treatment. The ORF15 sequence contained within this AAV vector, however, has ORF15 DNA sequence variations compared to the published sequence that are likely due to its unusual composition of repetitive purine nucleotides. This mutability is a concern for AAV vector production and safety when contemplating a human trial. In this study, we establish the safety profile of AAV-hIRBP-hRPGR and AAV-hGRK1-hRPGR vectors used in the initial canine proof-of-principle experiments by demonstrating hRPGR-ORF15 sequence stability during all phases of manipulation, from plasmid propagation to vector production to its stability in vivo after subretinal administration to animals. We also evaluate potential toxicity in vivo by investigating protein expression, retinal structure and function, and vector biodistribution. Expression of hRPGR is detected in the inner segments and synaptic terminals of photoreceptors and is restricted to the connecting cilium when the vector is further diluted. Treated eyes exhibit no toxicity as assessed by retinal histopathology, immunocytochemistry, optical coherence tomography, fundoscopy, electroretinogram, and vector biodistribution. Therefore, the hRPGR-ORF15 variant in our AAV vectors appears to be a more stable form than the endogenous hRPGR cDNA when propagated in vitro. Its safety profile presented here in combination with its proven efficacy supports future gene therapy clinical trials.


Subject(s)
Dependovirus/genetics , Eye Proteins/genetics , Genetic Therapy , Amino Acid Sequence , Animals , Base Sequence , Genetic Vectors , Humans , Male , Mice, Inbred C57BL , Molecular Sequence Data , Open Reading Frames , Retina/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...