Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Sci Rep ; 8(1): 4807, 2018 Mar 14.
Article in English | MEDLINE | ID: mdl-29540812

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

2.
PLoS One ; 12(11): e0188032, 2017.
Article in English | MEDLINE | ID: mdl-29131863

ABSTRACT

Adeno-associated virus (AAV) vectors are important gene delivery tools for the treatment of many recessively inherited retinal diseases. For example, a wild-type (WT) AAV5 vector can deliver a full-length Cnga3 (cyclic nucleotide-gated channel alpha-3) cDNA to target cells of the cone photoreceptor function loss 5 (cpfl5) mouse, a spontaneous animal model of achromatopsia with a Cnga3 mutation. Gene therapy restores cone-mediated function and blocks cone degeneration in the mice. However, since transgene expression delivered by an AAV vector shows relatively short-term effectiveness, this cannot be regarded as a very successful therapy. AAV2 and AAV8 vectors with capsid mutations have significantly enhanced transduction efficiency in retinas compared to WT AAV controls. In this study, AAV8 (Y447, 733F+T494V)-treated cpfl5 retinas showed greater preservation of short-term cone electroretinogram (ERG) responses than AAV8 (Y447, 733F)- or AAV2 (Y272, 444, 500, 730F+T491V)-mediated treatments. To explore the long-term rescue effect, AAV8 (Y447, 733F+T494V)-treated cpfl5 retinas were evaluated at 9 months following postnatal day 14 (P14) treatment. Rescued ERG responses in the cones of treated cpfl5 eyes decreased with increasing age, but still maintained more than 60% of the WT mouse responses at the oldest time point examined. Expression of CNGA3 and M/S-opsins was maintained in cone outer segments of the treated cpfl5 eyes and was equal to expression in age-matched WT retinas. Near-normal cone-mediated water maze behavior was observed in the treated cpfl5 mice. As these are the longest follow-up data reported thus far, AAV8 with capsid Y-F and T-V mutations may be one of the most effective AAV vectors for long-term treatment in a naturally occurring mouse model of CNGA3 achromatopsia.


Subject(s)
Capsid , Color Vision Defects/therapy , Cyclic Nucleotide-Gated Cation Channels/genetics , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors , Mutation , Retina/physiopathology , Animals , Behavior, Animal , Color Vision Defects/genetics , Color Vision Defects/physiopathology , Electroretinography , Genetic Therapy , Mice
3.
Sci Rep ; 7(1): 6690, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28751656

ABSTRACT

Cones are responsible for daylight, central, high acuity and color vision. Three proteins found in human cones, i.e. long-wavelength (L)-, middle-wavelength (M)-, and short-wavelength sensitive (S)-opsins, are responsible for red, green and blue color recognition, respectively. Human blue cone monochromacy (BCM) is characterized by functional loss of both L- and M-cone opsins due to mutations in the OPN1LW/OPN1MW gene cluster on the X chromosome. BCM patients, who rely on their vision from only S-cones and rods, suffer severely reduced visual acuity and impaired color vision. Recent studies show that there is sufficient cone structure remaining in the central fovea of BCM patients to consider AAV-mediated gene augmentation therapy. In contrast, mouse retina has only two opsins, S-opsin and M-opsin, but no L-opsin. We generated an M-opsin knockout mouse (Opn1mw -/-) expressing only S-opsin as a model for human BCM. We show that recombinant M-opsin delivered by AAV5 vectors rescues M-cone function in Opn1mw -/- mice. We also show that AAV delivered M-opsin localizes in the dorsal cone outer segments, and co-localizes with S-opsin in the ventral retina. Our study demonstrates that cones without M-opsin remain viable and respond to gene augmentation therapy, thereby providing proof-of-concept for cone function restoration in BCM patients.


Subject(s)
Color Vision Defects/genetics , Color Vision Defects/therapy , Genetic Therapy , Animals , Dependovirus/metabolism , Disease Models, Animal , Electroretinography , Humans , Mice , Mice, Knockout , Opsins/genetics , Opsins/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology
4.
J Ophthalmol ; 2017: 9721362, 2017.
Article in English | MEDLINE | ID: mdl-28168050

ABSTRACT

The retinal degeneration 11 (rd11) mouse is a new animal model with rapid photoreceptor degeneration. The long-term efficacy of gene therapy has a direct relationship with the onset of photoreceptor degeneration or apoptosis, whereas the degeneration or apoptosis patterns of photoreceptors are still unclear in rd11 mice. The distribution patterns of cone function-related L- and S-opsin were examined by immunofluorescence staining, and the apoptosis was performed by TUNEL assay in rd11 mice. The expression pattern of L-opsin or S-opsin in rd11 retina at postnatal day (P) 14 was similar to the pattern observed in wildtype retina. With increasing age, the expression of L-opsin and S-opsin, especially S-opsin, decreased significantly in rd11 mice. The degeneration of L-opsin began around the optic nerve and expanded to the periphery of the retina, from the ventral/nasal to dorsal/temporal retina, whereas the expression of S-opsin gradually decreased from the dorsal/temporal to ventral/nasal retina. Apoptotic signal appeared at P14 and was strongest at P28 of rd11 mice. The key genes associated with apoptosis confirmed those changes. These indicated that the degeneration and apoptosis of cone photoreceptors began at P14 of rd11 mice, which was a key point for gene therapy.

5.
PLoS One ; 11(5): e0156542, 2016.
Article in English | MEDLINE | ID: mdl-27228218

ABSTRACT

Lysophosphatidylcholine acyltransferase 1 (LPCAT1) is necessary for photoreceptors to generate an important lipid component of their membranes. The absence of LPCAT1 results in early and rapid rod and cone degeneration. Retinal degeneration 11 (rd11) mice carry a mutation in the Lpcat1 gene, and are an excellent model of early-onset rapid retinal degeneration (RD). To date, no reports have documented gene therapy administration in the rd11 mouse model at different ages. In this study, the AAV8 (Y733F)-smCBA-Lpcat1 vector was subretinally injected at postnatal day (P) 10, 14, 18, or 22. Four months after injection, immunohistochemistry and analysis of retinal morphology showed that treatment at P10 rescued about 82% of the wild-type retinal thickness. However, the diffusion of the vector and the resulting rescue were limited to an area around the injection site that was only 31% of the total retinal area. Injection at P14 resulted in vector diffusion that covered approximately 84% of the retina, and we found that gene therapy was more effective against RD when exposure to light was limited before and after treatment. We observed long-term preservation of electroretinogram (ERG) responses, and preservation of retinal structure, indicating that early treatment followed by limited light exposure can improve gene therapy effectiveness for the eyes of rd11 mice. Importantly, delayed treatment still partially preserved M-cones, but not S-cones, and M-cones in the rd11 retina appeared to have a longer window of opportunity for effective preservation with gene therapy. These results provide important information regarding the effects of subretinal gene therapy in the mouse model of LPCAT1-deficiency.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/biosynthesis , Genetic Therapy/methods , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/therapy , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Animals , Disease Models, Animal , Electroretinography , Humans , Mice , Mutation , Retinal Degeneration/epidemiology , Retinal Degeneration/genetics , Retinal Degeneration/physiopathology , Transduction, Genetic
7.
J Ophthalmol ; 2015: 250812, 2015.
Article in English | MEDLINE | ID: mdl-26137316

ABSTRACT

Retinal degenerative diseases are one of the important refractory ophthalmic diseases, featured with apoptosis of photoreceptor cells. Histone acetylation and deacetylation can regulate chromosome assembly, gene transcription, and posttranslational modification, which are regulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs), respectively. The histone deacetylase inhibitors (HDACis) have the ability to cause hyperacetylation of histone and nonhistone proteins, resulting in a variety of effects on cell proliferation, differentiation, anti-inflammation, and anti-apoptosis. Several HDACis have been approved for clinical trials to treat cancer. Studies have shown that HDACis have neuroprotective effects in nervous system damage. In this paper, we will summarize the neuroprotective effects of common HDACis in retinal degenerative diseases and make a prospect to the applications of HDACis in the treatment of retinal degenerative diseases in the future.

8.
Hum Mol Genet ; 24(13): 3699-707, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25855802

ABSTRACT

The CNGA3(-/-)/Nrl(-/-) mouse is a cone-dominant model with Cnga3 channel deficiency, which partially mimics the all cone foveal structure of human achromatopsia 2 with CNGA3 mutations. Although subretinal (SR) AAV vector administration can transfect retinal cells efficiently, the injection-induced retinal detachment can cause retinal damage, particularly when SR vector bleb includes the fovea. We therefore explored whether cone function-structure could be rescued in CNGA3(-/-)/Nrl(-/-) mice by intravitreal (IVit) delivery of tyrosine to phenylalanine (Y-F) capsid mutant AAV8. We find that AAV-mediated CNGA3 expression can restore cone function and rescue structure following IVit delivery of AAV8 (Y447, 733F) vector. Rescue was assessed by restoration of the cone-mediated electroretinogram (ERG), optomotor responses, and cone opsin immunohistochemistry. Demonstration of gene therapy in a cone-dominant mouse model by IVit delivery provides a potential alternative vector delivery mode for safely transducing foveal cones in achromatopsia patients and in other human retinal diseases affecting foveal function.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Color Vision Defects/genetics , Color Vision Defects/therapy , Cyclic Nucleotide-Gated Cation Channels/genetics , Eye Proteins/genetics , Genetic Therapy , Retinal Cone Photoreceptor Cells/physiology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Color Vision Defects/metabolism , Color Vision Defects/physiopathology , Cyclic Nucleotide-Gated Cation Channels/metabolism , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Eye Proteins/metabolism , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
PLoS One ; 10(2): e0117570, 2015.
Article in English | MEDLINE | ID: mdl-25706871

ABSTRACT

Early studies on Rpe65 knockout mice reported that remaining visual function was attributable to cone function. However, this finding has been challenged more and more as time has passed. Electroretinograms (ERGs) showed that rd12 mice, a spontaneous animal model of RPE65 Leber's congenital amaurosis, had sizeable photopic responses. Unfortunately, the recorded ERG waveform was difficult to interpret because of a remarkably delayed peak-time, which resembles a rod response more than a cone response. Here, we compare flicker ERGs in animals with normal rod and cone function (C57BL/6J mice), pure rod function (cpfl5 mice), and pure cone function (Rho(-/-) mice) under different adaptation levels and stimulus intensities. These responses were then compared with those obtained from rd12 mice. Our results showed that normal rods respond to low frequency flicker (5 and 15 Hz) and that normal cones respond to both low and high frequency flicker (5-35 Hz). As was seen in cpfl5 mice, rd12 mice had recordable responses to low frequency flicker (5 and 15Hz), but not to high frequency flicker (25 and 35 Hz). We hypothesize that abnormal rods may be the source of residual vision in rd12 mice, which is proved correct here with double mutant rd12mice. In this study, we show, for the first time, that frequency-response ERGs can effectively distinguish cone- and rod-driven responses in the rd12 mouse. It is another simple and valid method for evaluating the respective contributions of retinal rods and cones.


Subject(s)
Leber Congenital Amaurosis/physiopathology , Retinal Cone Photoreceptor Cells/physiology , Retinal Degeneration/physiopathology , Animals , Disease Models, Animal , Electroretinography , Mice , Mice, Knockout , cis-trans-Isomerases/genetics , rho GTP-Binding Proteins/genetics
10.
Invest Ophthalmol Vis Sci ; 55(3): 1724-34, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24557352

ABSTRACT

PURPOSE: The retinal degeneration 11 (rd11) mouse is a newly discovered, naturally occurring animal model with early photoreceptor dysfunction and rapid rod photoreceptor degeneration followed by cone degeneration. The rd11 mice carry a spontaneous mutation in the lysophosphatidylcholine acyltransferase 1 (Lpcat1) gene. Here, we evaluate whether gene replacement therapy using the fast-acting tyrosine-capsid mutant AAV8 (Y733F) can arrest retinal degeneration and restore retinal function in this model. METHODS: The AAV8 (Y733F)-smCBA-Lpcat1 was delivered subretinally to postnatal day 14 (P14) rd11 mice in one eye only. At 10 weeks after injection, treated rd11 mice were examined by visually-guided behavior, electroretinography (ERG) and spectral domain optical coherence tomography (SD-OCT), and then killed for morphologic and biochemical examination. RESULTS: Substantial scotopic and photopic ERG signals were maintained in treated rd11 eyes, whereas untreated eyes in the same animals showed extinguished signals. The SD-OCT (in vivo) and light microscopy (in vitro) showed a substantial preservation of the outer nuclear layer in most parts of the treated retina only. Almost wild-type LPCAT1 expression in photoreceptors with strong rod rhodopsin and M/S cone opsin staining, and normal visually-guided water maze behavioral performances were observed in treated rd11 mice. CONCLUSIONS: The results demonstrate that the tyrosine-capsid mutant AAV8 (Y733F) vector is effective for treating rapidly degenerating models of retinal degeneration and, moreover, is more therapeutically effective than AAV2 (Y444, 500, 730F) vector with the same promoter-cDNA payload. To our knowledge, this is the first demonstration of phenotypic rescue by gene therapy in an animal model of retinal degeneration caused by Lpcat1 mutation.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/genetics , DNA/genetics , Genetic Therapy/methods , Mutation , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/therapy , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Animals , Blotting, Western , DNA Mutational Analysis , Disease Models, Animal , Electroretinography , Genetic Vectors , Immunohistochemistry , Mice , Mice, Inbred C57BL , Retinal Degeneration/genetics , Retinal Degeneration/physiopathology , Tomography, Optical Coherence
11.
Zhonghua Yan Ke Za Zhi ; 49(6): 521-5, 2013 Jun.
Article in Chinese | MEDLINE | ID: mdl-24119965

ABSTRACT

OBJECTIVE: To investigate the distribution and biological roles of voltage-dependent calcium channel (VDCC) α1F subunit in murine retina. METHODS: Experimental study.α1F(-/-) (homozygous mutant) mice (n = 35) and α1F(+/+) (wild type) mice (n = 35) were used in this study. Immunohistochemistry was performed to determine the expression of VDCC α1F subunit in the mouse retina. Retinae in α1F(-/-) mice and age-matched control mice at 3, 6, 9, 14-day and 3-month after birth were paraffin embedded, sectioned and HE stained, and full-field electroretinogram (ERG) were also recorded at these time points.Statistics were based on independent samples t-test. RESULTS: (1) α1F subunit was absent in α1F(-/-) mice retina. But in α1F(+/+) mice retina, α1F subunit was expressed most strongly in the outer plexiform layer (OPL), less in the inner plexiform layer (IPL) and ganglion cell layer (GCL). (2) OPL thickness in the subunit deficient mice gradually reduced after birth and lost at adult age. (3) In dark-adapted ERGs,standard response showed that the b-wave amplitude of α1F(-/-) mice [(163.8 ± 26.7) µV] significantly decreased compared with that of α1F(+/+) mice [(408.4 ± 54.5) µV] (t = -9.017, P = 0.000), whereas the a-wave amplitude of α1F(-/-) group [(208.2 ± 27.3) µV] was similar to that of control group [(196.0 ± 24.2) µV] (t = 0.748, P = 0.476). CONCLUSION: This study demonstrates that the lack of VDCC α1F subunit affect the structure and function in the OPL of the murine retina.


Subject(s)
Calcium Channels, L-Type/genetics , Retina/pathology , Retina/physiopathology , Animals , Electroretinography , Homozygote , Mice , Mice, Knockout , Mutation
12.
Hum Gene Ther Clin Dev ; 24(1): 23-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23692380

ABSTRACT

Abstract Proof of concept for MERTK gene replacement therapy has been demonstrated using different viral vectors in the Royal College of Surgeon (RCS) rat, a well characterized model of recessive retinitis pigmentosa that contains a mutation in the Mertk gene. MERTK plays a key role in renewal of photoreceptor outer segments (OS) by phagocytosis of shed OS tips. Mutations in MERTK cause impaired phagocytic activity and accumulation of OS debris in the interphotoreceptor space that ultimately leads to photoreceptor cell death. In the present study, we conducted a series of preclinical potency and GLP-compliant safety evaluations of an adeno-associated virus type 2 (AAV2) vector expressing human MERTK cDNA driven by the retinal pigment epithelium-specific, VMD2 promoter. We demonstrate the potency of the vector in RCS rats by improved electroretinogram (ERG) responses in treated eyes compared with contralateral untreated controls. Toxicology and biodistribution studies were performed in Sprague-Dawley (SD) rats injected with two different doses of AAV vectors and buffer control. Delivery of vector in SD rats did not result in a change in ERG amplitudes of rod and cone responses relative to balanced salt solution control-injected eyes, indicating that administration of AAV vector did not adversely affect normal retinal function. In vivo fundoscopic analysis and postmortem retinal morphology of the vector-injected eyes were normal compared with controls. Evaluation of blood smears showed the lack of transformed cells in the treated eyes. All injected eyes and day 1 blood samples were positive for vector genomes, and all peripheral tissues were negative. Our results demonstrate the potency and safety of the AAV2-VMD2-hMERTK vector in animal models tested. A GMP vector has been manufactured and is presently in clinical trial.


Subject(s)
Dependovirus/genetics , Genetic Vectors/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Retinitis Pigmentosa/therapy , Animals , Bestrophins , Chloride Channels/genetics , Disease Models, Animal , Drug Evaluation, Preclinical , Eye Proteins/genetics , Female , Genetic Therapy , Genetic Vectors/genetics , Humans , Male , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/genetics , Retina/pathology , Retinitis Pigmentosa/pathology , Tissue Distribution , c-Mer Tyrosine Kinase
13.
Invest Ophthalmol Vis Sci ; 54(3): 1988-97, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23425697

ABSTRACT

PURPOSE: To elucidate the underlying pathologic mechanism of congenital stationary night blindness (CSNB) by examining the characteristics of electrical signal transmission within the inner retinal circuit after Cacna1f gene mutation. METHODS: Retinas isolated from the spontaneous Cacna1f mutant rats or wild-type rats were placed into a recording chamber, with the ganglion cell layer facing the biochip electrode array. The light-driven responses of the retinal ganglion cells (RCGs) were recorded using a multielectrode array (MEA) system. In the electrical stimulus cases, charge-balanced biphasic current pulse trains were generated and applied to the central electrode of MEA to stimulate the RCGs. Chemical compounds were bath-applied through an active perfusion system. The acquired data were further analyzed off-line. RESULTS: Typical electrical responses were successfully recorded in the retinas of both wild-type rats and Cacna1f gene mutated rats. In the Cacna1f mutant retinas, the amplitude of the light-induced a-wave was decreased, paralleling the vanished b-wave. The responsive a-wave was not blocked by the application of 100 µM 2-amino-4-phosphobutyric acid. The increased spontaneous firing rate and the decreased robustness of light-driven signaling reflected a loss in the ability of ganglion cells to encode visual signals reliably and economically. Moreover, the ON pathway is somehow disconnected from ganglion cells, whereas OFF pathways may be preferentially selected by the CSNB retinas. In the electrical stimulus cases, the long-latency responses of RGCs evoked by the indirect synaptic inputs from outer layers of retina were weaker in the CSNB rats compared with that of SD rats. CONCLUSIONS: Using MEA recording, we provide evidences of functional changes for visual signal pathway plasticity in the Cacna1f mutated retinas. Our results suggest that the dysfunctions in photoreceptor neurotransmitter release and the loss of signaling efficiency both occur during CSNB, and the latter is possibly reversible.


Subject(s)
Calcium Channels/genetics , DNA/genetics , Eye Diseases, Hereditary/genetics , Genetic Diseases, X-Linked/genetics , Mutation , Myopia/genetics , Night Blindness/genetics , Retina/physiopathology , Visual Pathways/physiology , Animals , Calcium Channels/metabolism , DNA Mutational Analysis , Disease Models, Animal , Electroretinography , Eye Diseases, Hereditary/metabolism , Eye Diseases, Hereditary/physiopathology , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/physiopathology , Myopia/metabolism , Myopia/physiopathology , Night Blindness/metabolism , Night Blindness/physiopathology , Rats , Rats, Mutant Strains , Retina/metabolism , Retina/pathology , Signal Transduction/genetics , Synaptic Transmission/genetics
14.
Zhonghua Yan Ke Za Zhi ; 48(8): 755-8, 2012 Aug.
Article in Chinese | MEDLINE | ID: mdl-23141518

ABSTRACT

Achromatopsia is an early onset retinal dystrophy that causes severe visual impairment. To date, four genes have been found to be implicated in achromatopsia-associated mutations: guanine nucleotide-binding protein (GNAT2), cyclic nucleotide-gated channel alpha-3 (CNGA3), cyclic nucleotide-gated channel beta-3 (CNGB3) and phosphodiesterase 6C (PDE6C). Even with early onset, the slow progress and the good responses to gene therapy in animal models render achromatopsia a very attractive candidate for human gene therapy after the successful of the Phase I clinical trials of Leber's congenital amaurosis. With the development of molecular genetics and the therapeutic gene replacement technology, the adeno-associated viral (AAV) vector-mediated gene therapy for achromatopsia in the preclinical animal experiments achieved encouraging progress in the past years. This article briefly reviews the recent research achievements of achromatopsia with gene therapy.


Subject(s)
Color Vision Defects/therapy , Genetic Therapy , Humans
15.
PLoS One ; 7(4): e35250, 2012.
Article in English | MEDLINE | ID: mdl-22509403

ABSTRACT

Achromatopsia is a rare autosomal recessive disorder which shows color blindness, severely impaired visual acuity, and extreme sensitivity to bright light. Mutations in the alpha subunits of the cone cyclic nucleotide-gated channels (CNGA3) are responsible for about 1/4 of achromatopsia in the U.S. and Europe. Here, we test whether gene replacement therapy using an AAV5 vector could restore cone-mediated function and arrest cone degeneration in the cpfl5 mouse, a naturally occurring mouse model of achromatopsia with a CNGA3 mutation. We show that gene therapy leads to significant rescue of cone-mediated ERGs, normal visual acuities and contrast sensitivities. Normal expression and outer segment localization of both M- and S-opsins were maintained in treated retinas. The therapeutic effect of treatment lasted for at least 5 months post-injection. This study is the first demonstration of substantial, relatively long-term restoration of cone-mediated light responsiveness and visual behavior in a naturally occurring mouse model of CNGA3 achromatopsia. The results provide the foundation for development of an AAV5-based gene therapy trial for human CNGA3 achromatopsia.


Subject(s)
Color Vision Defects/genetics , Color Vision Defects/therapy , Cyclic Nucleotide-Gated Cation Channels/genetics , Genetic Therapy , Animals , Cyclic Nucleotide-Gated Cation Channels/metabolism , Dependovirus , Disease Models, Animal , Electroretinography , Gene Expression Regulation , Genetic Vectors , Humans , Mice , Mutation , Opsins/genetics , Opsins/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology
16.
Neurobiol Aging ; 33(2): 433.e1-10, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21397984

ABSTRACT

To test the effects of adeno-associated virus encoding sFLT01 (AAV5.sFLT01) on the retinal lesions in Ccl2(-/-)/Cx3cr1(-/-) mice, a model for age-related macular degeneration (AMD), AAV5.sFLT01 was injected into the subretinal space of the right eyes and the left eyes served as controls. Histology found no retinal toxicity due to the treatment after 3 months. The treated eyes showed lesion arrest compared with lesion progression in the left eyes by fundus monitoring monthly and histological evaluation 3 months after treatment. Retinal ultrastructure showed fewer lipofuscin and better preserved photoreceptors after the treatment. A2E, a major component of lipofuscin, was lower in the treated eyes than in the control eyes. Molecular analysis showed that AAV5.sFLT01 lowered retinal extracellular signal-regulated kinase (ERK) phosphorylation and inducible nitric oxide synthetase expression, which suggested the involvement of reactive nitrogen species in the retinal lesions of Ccl2(-/-)/Cx3cr1(-/-). We concluded that local delivery of AAV5.sFLT01 can stabilize retinal lesions in Ccl2(-/-)/Cx3cr1(-/-) mice. The findings provide further support for the potential beneficial effects of sFLT01 gene therapy for age-related macular degeneration.


Subject(s)
Adenoviridae/genetics , Macular Degeneration/metabolism , Macular Degeneration/therapy , Transfection/methods , Vascular Endothelial Growth Factor Receptor-1/therapeutic use , Animals , CX3C Chemokine Receptor 1 , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Genetic Vectors , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics
17.
Arch Ophthalmol ; 130(1): 9-24, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21911650

ABSTRACT

OBJECTIVE: To determine the safety and efficacy of subretinal gene therapy in the RPE65 form of Leber congenital amaurosis using recombinant adeno-associated virus 2 (rAAV2) carrying the RPE65 gene. DESIGN: Open-label, dose-escalation phase I study of 15 patients (range, 11-30 years of age) evaluated after subretinal injection of the rAAV2- RPE65 vector into the worse-functioning eye. Five cohorts represented 4 dose levels and 2 different injection strategies. MAIN OUTCOME MEASURES: Primary outcomes were systemic and ocular safety. Secondary outcomes assayed visual function with dark-adapted full-field sensitivity testing and visual acuity with Early Treatment Diabetic Retinopathy Study charts. Further assays included immune responses to the vector, static visual fields, pupillometry, mobility performance, and optical coherence tomography. RESULTS: No systemic toxicity was detected; ocular adverse events were related to surgery. Visual function improved in all patients to different degrees; improvements were localized to treated areas. Cone and rod sensitivities increased significantly in the study eyes but not in the control eyes. Minor acuity improvements were recorded in many study and control eyes. Major acuity improvements occurred in study eyes with the lowest entry acuities and parafoveal fixation loci treated with subretinal injections. Other patients with better foveal structure lost retinal thickness and acuity after subfoveal injections. CONCLUSIONS: Gene therapy for Leber congenital amaurosis caused by RPE65 mutations is sufficiently safe and substantially efficacious in the extrafoveal retina. There is no benefit and some risk in treating the fovea. No evidence of age-dependent effects was found. Our results point to specific treatment strategies for subsequent phases. APPLICATION TO CLINICAL PRACTICE: Gene therapy for inherited retinal disease has the potential to become a future part of clinical practice. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00481546.


Subject(s)
Carrier Proteins/genetics , Dependovirus/genetics , Eye Proteins/genetics , Genetic Therapy/methods , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/therapy , Mutation , Adolescent , Adult , Child , Female , Follow-Up Studies , Genetic Therapy/adverse effects , Genetic Vectors , Humans , Injections, Intraocular , Leber Congenital Amaurosis/physiopathology , Male , Photic Stimulation , Photoreceptor Cells, Vertebrate/physiology , Psychomotor Performance/physiology , Pupil/physiology , Tomography, Optical Coherence , Treatment Outcome , Visual Acuity/physiology , Visual Fields/physiology , Young Adult , cis-trans-Isomerases
18.
Zhonghua Yan Ke Za Zhi ; 47(1): 83-7, 2011 Jan.
Article in Chinese | MEDLINE | ID: mdl-21418933

ABSTRACT

Leber congenital amaurosis (LCA) is an early onset retinal dystrophy that causes severe visual impairment. With the development of molecular genetics and the therapeutic gene replacement technology, the adeno-associated viral (AAV) vector-mediated gene therapy for LCA achieved encouraging progress in the past decade. The success of the Phase I clinical trials of human RPE65 gene therapy for LCA II patients makes it a pioneer in the field of retinal gene therapy and brings light to the cure of other hereditary retinopathy. This article briefly reviews the recent developments in the preclinical animal experiments and Phase I clinical trials for LCA.


Subject(s)
Genetic Therapy , Leber Congenital Amaurosis/therapy , Clinical Trials, Phase I as Topic , Humans
19.
Mol Ther ; 19(2): 234-42, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21139570

ABSTRACT

The retinal degeneration 10 (rd10) mouse is a well-characterized model of autosomal recessive retinitis pigmentosa (RP), which carries a spontaneous mutation in the ß subunit of rod cGMP-phosphodiesterase (PDEß). Rd10 mouse exhibits photoreceptor dysfunction and rapid rod photoreceptor degeneration followed by cone degeneration and remodeling of the inner retina. Here, we evaluate whether gene replacement using the fast-acting tyrosine-capsid mutant AAV8 (Y733F) can provide long-term therapy in this model. AAV8 (Y733F)-smCBA-PDEß was subretinally delivered to postnatal day 14 (P14) rd10 mice in one eye only. Six months after injection, spectral domain optical coherence tomography (SD-OCT), electroretinogram (ERG), optomotor behavior tests, and immunohistochemistry showed that AAV8 (Y733F)-mediated PDEß expression restored retinal function and visual behavior and preserved retinal structure in treated rd10 eyes for at least 6 months. This is the first demonstration of long-term phenotypic rescue by gene therapy in an animal model of PDEß-RP. It is also the first example of tyrosine-capsid mutant AAV8 (Y733F)-mediated correction of a retinal phenotype. These results lay the groundwork for the development of PDEß-RP gene therapy trial and suggest that tyrosine-capsid mutant AAV vectors may be effective for treating other rapidly degenerating models of retinal degeneration.


Subject(s)
Capsid/metabolism , Dependovirus/genetics , Genetic Vectors/genetics , Retinitis Pigmentosa/therapy , Animals , Blotting, Western , Disease Models, Animal , Electroretinography , Genetic Therapy , Immunohistochemistry , Mice , Mice, Inbred C57BL , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology
20.
Invest Ophthalmol Vis Sci ; 52(1): 7-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21169527

ABSTRACT

PURPOSE: RPE65 function is necessary in the retinal pigment epithelium (RPE) to generate chromophore for all opsins. Its absence results in vision loss and rapid cone degeneration. Recent Leber congenital amaurosis type 2 (LCA with RPE65 mutations) phase I clinical trials demonstrated restoration of vision on RPE65 gene transfer into RPE cells overlying cones. In the rd12 mouse, a naturally occurring model of RPE65-LCA early cone degeneration was observed; however, some peripheral M-cones remained. A prior study showed that AAV-mediated RPE65 expression can prevent early cone degeneration. The present study was conducted to test whether the remaining cones in older rd12 mice can be rescued. METHODS: Subretinal treatment with the scAAV5-smCBA-hRPE65 vector was initiated at postnatal day (P)14 and P90. After 2 months, electroretinograms were recorded, and cone morphology was analyzed by using cone-specific peanut agglutinin and cone opsin-specific antibodies. RESULTS: Cone degeneration started centrally and spread ventrally, with cells losing cone-opsin staining before that for the PNA-lectin-positive cone sheath. Gene therapy starting at P14 resulted in almost wild-type M- and S-cone function and morphology. Delaying gene-replacement rescued the remaining M-cones, and most important, more M-cone opsin-positive cells were identified than were present at the onset of gene therapy, suggesting that opsin expression could be reinitiated in cells with cone sheaths. CONCLUSIONS: The results support and extend those of the previous study that gene therapy can stop early cone degeneration, and, more important, they provide proof that delayed treatment can restore the function and morphology of the remaining cones. These results have important implications for the ongoing LCA2 clinical trials.


Subject(s)
Carrier Proteins/genetics , Dependovirus/genetics , Disease Models, Animal , Eye Proteins/genetics , Genetic Therapy , Retinal Cone Photoreceptor Cells/physiology , Animals , Cone Opsins/metabolism , Electroretinography , Fluorescent Antibody Technique, Indirect , Genetic Vectors , Green Fluorescent Proteins/genetics , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/physiopathology , Leber Congenital Amaurosis/therapy , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , cis-trans-Isomerases
SELECTION OF CITATIONS
SEARCH DETAIL
...