Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Cells ; 13(5)2024 Feb 25.
Article in English | MEDLINE | ID: mdl-38474360

ABSTRACT

Embolism, hyperglycemia, high intraocular pressure-induced increased reactive oxygen species (ROS) production, and microglial activation result in endothelial/retinal ganglion cell death. Here, we conducted in vitro and in vivo ischemia/reperfusion (I/R) efficacy studies of a hybrid antioxidant-nitric oxide donor small molecule, SA-10, to assess its therapeutic potential for ocular stroke. METHODS: To induce I/R injury and inflammation, we subjected R28 and primary microglial cells to oxygen glucose deprivation (OGD) for 6 h in vitro or treated these cells with a cocktail of TNF-α, IL-1ß and IFN-γ for 1 h, followed by the addition of SA-10 (10 µM). Inhibition of microglial activation, ROS scavenging, cytoprotective and anti-inflammatory activities were measured. In vivo I/R-injured mouse retinas were treated with either PBS or SA-10 (2%) intravitreally, and pattern electroretinogram (ERG), spectral-domain optical coherence tomography, flash ERG and retinal immunocytochemistry were performed. RESULTS: SA-10 significantly inhibited microglial activation and inflammation in vitro. Compared to the control, the compound SA-10 significantly attenuated cell death in both microglia (43% vs. 13%) and R28 cells (52% vs. 17%), decreased ROS (38% vs. 68%) production in retinal microglia cells, preserved neural retinal function and increased SOD1 in mouse eyes. CONCLUSION: SA-10 is protective to retinal neurons by decreasing oxidative stress and inflammatory cytokines.


Subject(s)
Reperfusion Injury , Retinal Ganglion Cells , Mice , Animals , Reactive Oxygen Species/metabolism , Retinal Ganglion Cells/metabolism , Reperfusion Injury/metabolism , Ischemia/metabolism , Anti-Inflammatory Agents/therapeutic use , Inflammation/metabolism , Reperfusion
2.
Article in English | MEDLINE | ID: mdl-38464735

ABSTRACT

Glaucoma is a chronic and progressive eye disease, commonly associated with elevated intraocular pressure (IOP) and characterized by optic nerve degeneration, cupping of the optic disc, and loss of retinal ganglion cells (RGCs). The pathological changes in glaucoma are triggered by multiple mechanisms and both mechanical effects and vascular factors are thought to contribute to the etiology of glaucoma. Various studies have shown that endothelin-1 (ET-1), a vasoactive peptide, acting through its G protein coupled receptors, ETA and ETB, plays a pathophysiologic role in glaucoma. However, the mechanisms by which ET-1 contribute to neurodegeneration remain to be completely understood. Our laboratory and others demonstrated that macitentan (MAC), a pan endothelin receptor antagonist, has neuroprotective effects in rodent models of IOP elevation. The current study aimed to determine if oral administration of a dual endothelin antagonist, macitentan, could promote neuroprotection in an acute model of intravitreal administration of ET-1. We demonstrate that vasoconstriction following the intravitreal administration of ET-1 was attenuated by dietary administration of the ETA/ETB dual receptor antagonist, macitentan (5 mg/kg body weight) in retired breeder Brown Norway rats. ET-1 intravitreal injection produced a 40% loss of RGCs, which was significantly lower in macitentan-treated rats. We also evaluated the expression levels of glial fibrillary acidic protein (GFAP) at 24 h and 7 days post intravitreal administration of ET-1 in Brown Norway rats as well as following ET-1 treatment in cultured human optic nerve head astrocytes. We observed that at the 24 h time point the expression levels of GFAP was upregulated (indicative of glial activation) following intravitreal ET-1 administration in both retina and optic nerve head regions. However, following macitentan administration for 7 days after intravitreal ET-1 administration, we observed an upregulation of GFAP expression, compared to untreated rats injected intravitreally with ET-1 alone. Macitentan treatment in ET-1 administered rats showed protection of RGC somas but was not able to preserve axonal integrity and functionality. The endothelin receptor antagonist, macitentan, has neuroprotective effects in the retinas of Brown Norway rats acting through different mechanisms, including enhancement of RGC survival and reduction of ET-1 mediated vasoconstriction.

3.
J Cell Mol Med ; 25(18): 8676-8686, 2021 09.
Article in English | MEDLINE | ID: mdl-34418301

ABSTRACT

Gene therapy of retinal diseases using recombinant adeno-associated virus (rAAV) vector-based delivery has shown clinical success, and clinical trials based on rAAV-based optogenetic therapies are currently in progress. Recently, we have developed multi-characteristic opsin (MCO), which has been shown to effectively re-photosensitize photoreceptor-degenerated retina in mice leading to vision restoration at ambient light environment. Here, we report the biodistribution of the rAAV2 carried MCO (vMCO-I) in live samples and post-mortem organs following intraocular delivery in wild-type dogs. Immunohistochemistry showed that the intravitreal injection of vMCO-I resulted in gene transduction in the inner nuclear layer (INL) but did not induce detectable inflammatory or immune reaction in the dog retina. Vector DNA analysis of live body wastes and body fluids such as saliva and nasal secretions using quantitative polymerase chain reaction (qPCR) showed no correlative increase of vector copy in nasal secretions or saliva, minimal increase of vector copy in urine in the low-dose group 13 weeks after injection and in the faeces of the high-dose group at 3-13 weeks after injection suggesting clearance of the virus vector via urine and faeces. Further analysis of vector DNA extracted from faeces using PCR showed no transgene after 3 weeks post-injection. Intravitreal injection of vMCO-I resulted in few sporadic off-target presences of the vector in the mesenteric lymph node, liver, spleen and testis. This study showed that intravitreal rAAV2-based delivery of MCO-I for retinal gene therapy is safe.


Subject(s)
Dependovirus/physiology , Genetic Therapy/methods , Retinal Diseases/therapy , Animals , Dogs , Female , Genetic Vectors , Male
4.
JAAD Case Rep ; 6(9): 868-870, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32875037
6.
Nature ; 581(7806): 83-88, 2020 05.
Article in English | MEDLINE | ID: mdl-32376950

ABSTRACT

Photoreceptor loss is the final common endpoint in most retinopathies that lead to irreversible blindness, and there are no effective treatments to restore vision1,2. Chemical reprogramming of fibroblasts offers an opportunity to reverse vision loss; however, the generation of sensory neuronal subtypes such as photoreceptors remains a challenge. Here we report that the administration of a set of five small molecules can chemically induce the transformation of fibroblasts into rod photoreceptor-like cells. The transplantation of these chemically induced photoreceptor-like cells (CiPCs) into the subretinal space of rod degeneration mice (homozygous for rd1, also known as Pde6b) leads to partial restoration of the pupil reflex and visual function. We show that mitonuclear communication is a key determining factor for the reprogramming of fibroblasts into CiPCs. Specifically, treatment with these five compounds leads to the translocation of AXIN2 to the mitochondria, which results in the production of reactive oxygen species, the activation of NF-κB and the upregulation of Ascl1. We anticipate that CiPCs could have therapeutic potential for restoring vision.


Subject(s)
Cellular Reprogramming/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Retinal Degeneration/therapy , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/transplantation , Vision, Ocular/drug effects , Animals , Axin Protein/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Disease Models, Animal , Flow Cytometry , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NF-kappa B/metabolism , Protein Transport/drug effects , Reactive Oxygen Species/metabolism , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/drug effects , Signal Transduction/drug effects , Vision, Ocular/physiology
7.
Am J Ophthalmol Case Rep ; 18: 100677, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32322747
8.
Invest Ophthalmol Vis Sci ; 60(8): 3064-3073, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31348824

ABSTRACT

Purpose: Determine the toxicity, bioavailability in the retina, and neuroprotective effects of a hybrid antioxidant-nitric oxide donor compound SA-2 against oxidative stress-induced retinal ganglion cell (RGC) death in neurodegenerative animal models. Methods: Optic nerve crush (ONC) and ischemia reperfusion (I/R) injury models were used in 12-week-old C57BL/6J mice to mimic conditions of glaucomatous neurodegeneration. Mice were treated intravitreally with either vehicle or SA-2. Retinal thickness was measured by spectral-domain optical coherence tomography (SD-OCT). The electroretinogram and pattern ERG (PERG) were used to assess retinal function. RGC survival was determined by counting RBPMS-positive RGCs and immunohistochemical analysis of superoxide dismutase 1 (SOD1) levels was carried out in the retina sections. Concentrations of SA-2 in the retina and choroid were determined using HPLC and MS. In addition, the direct effect of SA-2 treatment on RGC survival was assessed in ex vivo rat retinal explants under hypoxic (0.5% O2) conditions. Results: Compound SA-2 did not induce any appreciable change in retinal thickness, or in a- or b-wave amplitude in naive animals. SA-2 was found to be bioavailable in both the retina and choroid after a single intravitreal injection (2% wt/vol). An increase in SOD1 levels in the retina of mice subjected to ONC and SA-2 treatment, suggests an enhancement in antioxidant activity. SA-2 provided significant (P < 0.05) RGC protection in all three of the tested RGC injury models in rodents. PERG amplitudes were significantly higher in both I/R and ONC mouse eyes following SA-2 treatment (P ≤ 0.001) in comparison with the vehicle and control groups. Conclusions: Compound SA-2 was effective in preventing RGC death and loss of function in three different rodent models of acute RGC injury: ONC, I/R, and hypoxia.


Subject(s)
Neuroprotection/drug effects , Nitric Oxide Donors/pharmacokinetics , Oxidative Stress , Retinal Degeneration/drug therapy , Retinal Ganglion Cells/pathology , Animals , Cell Survival/drug effects , Disease Models, Animal , Electroretinography , Female , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Ganglion Cells/metabolism , Tomography, Optical Coherence
9.
Am J Pathol ; 188(9): 2087-2096, 2018 09.
Article in English | MEDLINE | ID: mdl-30126549

ABSTRACT

Uveitis is estimated to account for 10% of all cases of blindness in the United States, including 30,000 new cases of legal blindness each year. Intraocular and oral corticosteroids are the effective mainstay treatment, but they carry the risk of serious long-term ocular and systemic morbidity. New noncorticosteroid therapies with a favorable side effect profile are necessary for the treatment of chronic uveitis, given the paucity of existing treatment choices. We have previously demonstrated that Nutlin-3, a small-molecule inhibitor of murine double minute 2 (MDM2) homolog, suppresses pathologic retinal angiogenesis through a p53-dependent mechanism, but the noncanonical p53-independent functions have not been adequately elucidated. Herein, we demonstrate an unanticipated function of MDM2 inhibition, where Nutlin-3 potently abrogates lipopolysaccharide-induced ocular inflammation. Furthermore, we identified a mechanism by which transcription and translation of NF-κB is mediated by MDM2, independent of p53, in ocular inflammation. Small-molecule MDM2 inhibition is a novel noncorticosteroid strategy for inhibiting ocular inflammation, which may potentially benefit patients with chronic uveitis.


Subject(s)
Eye Diseases/prevention & control , Gene Expression Regulation/drug effects , Imidazoles/pharmacology , Inflammation/prevention & control , NF-kappa B/antagonists & inhibitors , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Eye Diseases/metabolism , Eye Diseases/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism
10.
ACS Nano ; 11(5): 4669-4685, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28463509

ABSTRACT

Age-related macular degeneration (AMD) is the foremost cause of irreversible blindness in people over the age of 65 especially in developing countries. Therefore, an exploration of effective and alternative therapeutic interventions is an unmet medical need. It has been established that oxidative stress plays a key role in the pathogenesis of AMD, and hence, neutralizing oxidative stress is an effective therapeutic strategy for treatment of this serious disorder. Owing to autoregenerative properties, nanoceria has been widely used as a nonenzymatic antioxidant in the treatment of oxidative stress related disorders. Yet, its potential clinical implementation has been greatly hampered by its poor water solubility and lack of reliable tracking methodologies/processes and hence poor absorption, distribution, and targeted delivery. The water solubility and surface engineering of a drug with biocompatible motifs are fundamental to pharmaceutical products and precision medicine. Here, we report an engineered water-soluble, biocompatible, trackable nanoceria with enriched antioxidant activity to scavenge intracellular reactive oxygen species (ROS). Experimental studies with in vitro and in vivo models demonstrated that this antioxidant is autoregenerative and more active in inhibiting laser-induced choroidal neovascularization by decreasing ROS-induced pro-angiogenic vascular endothelial growth factor (VEGF) expression, cumulative oxidative damage, and recruitment of endothelial precursor cells without exhibiting any toxicity. This advanced formulation may offer a superior therapeutic effect to deal with oxidative stress induced pathogeneses, such as AMD.


Subject(s)
Cerium/pharmacology , Oxidative Stress/drug effects , Animals , Antioxidants/pharmacology , Cerium/chemistry , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/prevention & control , Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Humans , Macular Degeneration/physiopathology , Macular Degeneration/therapy , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Vascular Endothelial Growth Factor A/metabolism
11.
Retina ; 36(12): 2304-2310, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27205892

ABSTRACT

PURPOSE: Failure of blood pressure (BP) to dip during sleep (nondipper pattern) is associated with cardiovascular disease and stroke. The prevalence and degree of nondipping and masked hypertension in patients with retinal vein occlusion (RVO), which is associated with stroke, has not been previously examined. METHODS: We measured clinic and 24-hour ambulatory BPs in 22 patients with RVO and 20 control participants without known eye disease matched by age and sex. Mean BP dipping, defined as the ratio of difference in mean awake and sleep systolic BPs to mean awake systolic BP, and masked and nocturnal hypertension were compared between groups. RESULTS: Mean 24-hour ambulatory BP was 144/79 mmHg among those with RVO and 136/77 mmHg among controls. Patients with RVO had an almost 2-fold higher prevalence of nondipping pattern (80.8% [95% confidence interval, 52.8-94.1] vs. 50.4% [95% confidence interval, 26.1-74.5]; P = 0.008). Average sleep systolic BP dip in patients with RVO was 6.1% versus 11.9% in controls (P = 0.004). More patients with RVO had masked hypertension by ambulatory BPs than controls (71% vs. 50%), but this difference was not statistically significant. CONCLUSION: Our data suggest an association between RVO and nondipper BP pattern. Ambulatory BP monitoring may be useful in the evaluation of patients with RVO by identifying those who may benefit from more aggressive BP control.


Subject(s)
Blood Pressure Monitoring, Ambulatory , Blood Pressure/physiology , Retinal Vein Occlusion/physiopathology , Aged , Case-Control Studies , Circadian Rhythm/physiology , Cross-Sectional Studies , Female , Humans , Hypertension/epidemiology , Hypertension/physiopathology , Male , Middle Aged , Prevalence
14.
Methods Mol Biol ; 1357: 395-401, 2016.
Article in English | MEDLINE | ID: mdl-25403469

ABSTRACT

Reprogramming fibroblasts into induced pluripotent stem cells (iPSC) remains a promising technique for cell replacement therapy. Diverse populations of somatic cells have been examined for their reprogramming potential. Recently, ocular ciliary body epithelial cells (CECs) have been reprogrammed with high reprogramming efficiency and single transcription factor reprogramming, making them an exciting candidate for cellular reprogramming strategies.


Subject(s)
Cell Culture Techniques/methods , Cellular Reprogramming Techniques/methods , Cellular Reprogramming , Ciliary Body/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , Cellular Reprogramming/drug effects , Culture Media/pharmacology , Epithelial Cells/cytology , Genetic Vectors/genetics , Induced Pluripotent Stem Cells/drug effects , Lentivirus/genetics , Mice , Recombinant Fusion Proteins/genetics , Spheroids, Cellular , Transcription Factors/genetics , Transcription Factors/physiology
15.
Molecules ; 20(11): 19690-8, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26528964

ABSTRACT

Tetracycline (TC) is a well-known broad spectrum antibiotic, which is effective against many Gram positive and Gram negative bacteria. Controlled release nanoparticle formulations of TC have been reported, and could be beneficial for application in the treatment of periodontitis and dental bone infections. Furthermore, TC-controlled transcriptional regulation systems (Tet-on and Tet-off) are useful for controlling transgene expression in vitro and in vivo for biomedical research purposes; controlled TC release systems could be useful here, as well. Mesoporous silica nanomaterials (MSNs) are widely studied for drug delivery applications; Mobile crystalline material 41 (MCM-41), a type of MSN, has a mesoporous structure with pores forming channels in a hexagonal fashion. We prepared 41 ± 4 and 406 ± 55 nm MCM-41 mesoporous silica nanoparticles and loaded TC for controlled dug release; TC content in the TC-MCM-41 nanoparticles was 18.7% and 17.7% w/w, respectively. Release of TC from TC-MCM-41 nanoparticles was then measured in phosphate-buffered saline (PBS), pH 7.2, at 37 °C over a period of 5 h. Most antibiotic was released from both over this observation period; however, the majority of TC was released over the first hour. Efficacy of the TC-MCM-41 nanoparticles was then shown to be superior to free TC against Escherichia coli (E. coli) in culture over a 24 h period, while blank nanoparticles had no effect.


Subject(s)
Escherichia coli/drug effects , Nanoparticles , Silicon Dioxide , Tetracycline/administration & dosage , Delayed-Action Preparations , Drug Carriers , Drug Delivery Systems , Microbial Sensitivity Tests , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Silicon Dioxide/chemistry
17.
Stem Cells Dev ; 23(24): 3065-71, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25027059

ABSTRACT

Somatic cells can be reprogrammed to induced pluripotent stem (iPS) cells by defined sets of transcription factors. We previously described reprogramming of monolayer-cultured adult mouse ciliary body epithelial (CE) cells by Oct4 and Klf4, but not with Oct4 alone. In this study, we report that Oct4 alone is sufficient to reprogram CE cells to iPS cells through sphere formation. Furthermore, we demonstrate that sphere formation induces a partial reprogramming state characterized by expression of retinal progenitor markers, upregulation of reprogramming transcription factors, such as Sall4 and Nanog, demethylation in the promoter regions of pluripotency associated genes, and mesenchymal to epithelial transition. The Oct4-iPS cells maintained normal karyotypes, expressed markers for pluripotent stem cells, and were capable of differentiating into derivatives of all three embryonic germ layers in vivo and in vitro. These findings suggest that sphere formation may render somatic cells more susceptible to reprogramming.


Subject(s)
Cellular Reprogramming , Ciliary Body/cytology , Epithelial Cells/cytology , Induced Pluripotent Stem Cells/cytology , Octamer Transcription Factor-3/metabolism , Animals , Epithelial Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Octamer Transcription Factor-3/genetics , Spheroids, Cellular
18.
Br J Ophthalmol ; 98(7): 984-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24671925

ABSTRACT

BACKGROUND: To study spatiotemporal in vivo changes in retinal morphology and quantify thickness of retinal layers in a mouse model of light-induced retinal degeneration using spectral domain optical coherence tomography (SD-OCT). METHODS: BALB/c mice were exposed to 5000 lux of constant light for 3 h. SD-OCT images were taken 3 h, 24 h, 3 days, 1 week and 1 month after light exposure and were compared with histology at the same time points. SD-OCT images were also taken at 0, 1 and 2 h after light exposure in order to analyse retinal changes at the earliest time points. The thickness of retinal layers was measured using the Bioptigen software InVivoVue Diver. RESULTS: SD-OCT demonstrated progressive outer retinal thinning. 3 h after light exposure, the outer nuclear layer converted from hyporeflective to hyper-reflective. At 24 h, outer retinal bands and nuclear layer demonstrated similar levels of hyper-reflectivity. Significant variations in outer retinal thickness, vitreous opacities and retinal detachments occurred within days of injury. Thinning of the retina was observed at 1 month after injury. It was also determined that outer nuclear layer changes precede photoreceptor segment structure disintegration and the greatest change in segment structure occurs between 1 and 2 h after light exposure. CONCLUSIONS: Longitudinal SD-OCT reveals intraretinal changes that cannot be observed by histopathology at early time points in the light injury model.


Subject(s)
Light/adverse effects , Photoreceptor Cells, Vertebrate/ultrastructure , Radiation Injuries, Experimental/pathology , Retina/radiation effects , Retinal Degeneration/pathology , Animals , Female , Mice , Mice, Inbred BALB C , Radiation Injuries, Experimental/etiology , Retinal Degeneration/etiology , Tomography, Optical Coherence
19.
J Clin Invest ; 123(10): 4170-81, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24018558

ABSTRACT

Neovascular age-related macular degeneration is a leading cause of irreversible vision loss in the Western world. Cytokine-targeted therapies (such as anti-vascular endothelial growth factor) are effective in treating pathologic ocular angiogenesis, but have not led to a durable effect and often require indefinite treatment. Here, we show that Nutlin-3, a small molecule antagonist of the E3 ubiquitin protein ligase MDM2, inhibited angiogenesis in several model systems. We found that a functional p53 pathway was essential for Nutlin-3-mediated retinal antiangiogenesis and disruption of the p53 transcriptional network abolished the antiangiogenic activity of Nutlin-3. Nutlin-3 did not inhibit established, mature blood vessels in the adult mouse retina, suggesting that only proliferating retinal vessels are sensitive to Nutlin-3. Furthermore, Nutlin-3 inhibited angiogenesis in nonretinal models such as the hind limb ischemia model. Our work demonstrates that Nutlin-3 functions through an antiproliferative pathway with conceivable advantages over existing cytokine-targeted antiangiogenesis therapies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Imidazoles/pharmacology , Neovascularization, Physiologic/drug effects , Piperazines/pharmacology , Retinal Vessels/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cell Proliferation , Cell Survival , Cells, Cultured , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Ischemia/drug therapy , Macular Degeneration/drug therapy , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Rats , Retinal Vessels/drug effects , Transcriptional Activation/drug effects , Tumor Suppressor Protein p53/genetics
20.
Mol Ther ; 21(12): 2205-16, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23939025

ABSTRACT

A drawback of gene therapy using adeno-associated virus (AAV) is the DNA packaging restriction of the viral capsid (<4.7 kb). Recent observations demonstrate oversized AAV genome transduction through an unknown mechanism. Herein, AAV production using an oversized reporter (6.2 kb) resulted in chloroform and DNase-resistant particles harboring distinct "fragment" AAV (fAAV) genomes (5.0, 2.4, and 1.6 kb). Fractionation experiments determined that only the larger "fragments" mediated transduction in vitro, and relatively efficient transduction was also demonstrated in the muscle, the eye, and the liver. In contrast with concatemerization-dependent large-gene delivery by split AAV, fAAV transduction is independent of the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) in vitro and in vivo while disproportionately reliant on the DNA strand-annealing protein Rad51C. Importantly, fAAV's unique dependence on DNA repair proteins, compared with intact AAV, strongly suggests that the majority of oversized AAV transduction is mediated by fragmented genomes. Although fAAV transduction is less efficient than intact AAV, it is enhanced fourfold in muscle and sevenfold in the retina compared with split AAV transduction. Furthermore, fAAV carrying codon-optimized therapeutic dysferlin cDNA in a 7.5 kb expression cassette restored dysferlin levels in a dystrophic model. Collectively, oversized AAV genome transduction requires unique DNA repair pathways and offers an alternative, more efficient strategy for large-gene therapy.


Subject(s)
Capsid/metabolism , DNA-Binding Proteins/genetics , Dependovirus/physiology , Genetic Therapy , Liver/virology , Muscles/virology , Rad51 Recombinase/genetics , Transduction, Genetic , Animals , CHO Cells , Cricetulus , DNA Packaging , DNA, Viral/genetics , DNA-Binding Proteins/metabolism , Dependovirus/genetics , Disease Models, Animal , Eye/virology , Genes, Reporter , Genetic Vectors , Genome, Viral , HEK293 Cells , HeLa Cells , Humans , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/virology , Nucleic Acid Conformation , Rad51 Recombinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...