Abstract
Blindness, although not life threatening, is a debilitating disorder for which few, if any treatments exist. Ocular gene therapies have the potential to profoundly improve the quality of life in patients with inherited retinal disease. As such, tremendous focus has been given to develop such therapies. Several factors make the eye an ideal organ for gene-replacement therapy including its accessibility, immune privilege, small size, compartmentalization, and the existence of a contralateral control. This review will provide a comprehensive summary of (i) existing gene therapy clinical trials for several genetic forms of blindness and (ii) preclinical efficacy and safety studies in a variety of animal models of retinal disease which demonstrate strong potential for clinical application. To be as comprehensive as possible, we include additional proof of concept studies using gene replacement, neurotrophic/neuroprotective, optogenetic, antiangiogenic, or antioxidative stress strategies as well as a description of the current challenges and future directions in the ocular gene therapy field to this review as a supplement.
Introduction
The eye is a complex sensory organ that has evolved to both promote survival and allow us an appreciation of the order and beauty of our surroundings. Rodieck wisely remarks in his prologue, “so immediate and powerful is seeing that our language gives this word additional connotations; to imagine, to comprehend, to regard, to perceive, to know from first-hand experience, to foresee.”1 Understandably, the loss of vision due to inherited or acquired retinal disease can affect one's life in significant and sometimes devastating ways. Over the past few decades, vision scientists have worked to delineate the underlying molecular events which contribute to these diseases. This knowledge, combined with thorough clinical characterization of patients has led to the development of gene-replacement strategies for numerous inherited and acquired retinal diseases. This review will summarize existing early-stage clinical trials for several forms of genetic blindness and a variety of preclinical studies in which gene transfer resulted in significant functional improvement and/or regeneration or stabilization of retinal structure in animal models of retinal disease. Focus will be given to the therapies which demonstrate the strongest potential for clinical application in the near future. Additional gene replacement, neurotrophic/neuroprotective, optogenetic, antiangiogenic, or antioxidative stress strategies as well as a description of the current challenges and future directions in the ocular gene therapy field are provided as a supplement (Supplementary Data).
In Clinical Trial
RPE65-Leber congenital amaurosis
RPE65-Leber congenital amaurosis (LCA2) is associated with mutations in RPE65 (retinal pigment epithelium-specific 65 kDa protein). RPE65 is almost exclusively expressed in the RPE and functions as the retinoid isomerase responsible for converting all-trans retinoid to 11-cis retinal during pigment regeneration.2,3,4 Improper functioning or absence of RPE65 results in a lack of 11-cis retinal production and an inability to efficiently form the visual pigments, rhodopsin and cone opsin. Concomitant accumulation of large amounts of all-trans-retinyl esters in the RPE is thought to promote photoreceptor degeneration. As in patients with LCA2, the absence of isomerase activity in the RPE65 knock-out mouse, mutant knock-in mouse and naturally occurring RPE65 mutant mouse and dog models results in retinal degeneration and severe visual impairment. The natural histories of patients with LCA2 and the aforementioned animal models as well as a summary of experiments that paved the way for clinical treatment of LCA2 has been recently reviewed by Cideciyan.5
Despite its rarity (less than 1 in 1,000,000 live births affected), because of its early onset and the availability of multiple animal models, a tremendous amount of attention has been focused on developing a gene-based therapy for LCA2. Results published in 2001 describing successful gene therapy using a recombinant adeno-associated virus (AAV2) vector containing RPE65 cDNA to treat three RPE65 mutant Briard dogs generated much excitement and optimism in the field.6 In this study, subretinal delivery of a recombinant AAV2 vector carrying the canine RPE65 gene, under the control of the hybrid cytomegalovirus/chicken β-actin (CBA) promoter, resulted in substantial improvements in visual function out to 3 months postinjection, as assessed by an improvement in the electroretinogram (ERG).6 Subsequent follow-up studies by a number of groups would confirm and extend these results, as well as determine that subretinally delivered AAV1, AAV4, and AAV5-mediated RPE65 expression were also capable of restoring some level of function.7,8,9,10,11,12,13 Most encouraging were the findings that gains in visual function remained stable over time.8,9,13 Additional tests revealed that visually guided behavior was restored to treated dogs suggesting that retinal responses were being propagated to the visual processing centers of the brain.6,7,12 Cortical responses were assessed using functional magnetic resonance imaging and, as expected, were dramatically improved as a consequence of AAV2-RPE65 treatment.14
In addition to the dog studies, gene-replacement studies utilizing AAV, lentiviral and adenoviral vectors carrying RPE65 were carried out in murine models of LCA2. The first LCA2 mouse model used in gene-replacement studies was the RPE65 knock-out (Rpe65−/−) mouse generated and characterized by the Redmond laboratory.15 Subretinal injection of AAV1-RPE65 resulted in measurable improvements in ERG responses in Rpe65−/− mice when treated at a variety of ages, as early as in-utero, with efficacy followed out to as late as 24 months of age.16,17 Subretinal delivery of adenoviral-RPE65 in RPE65−/− mice reconstituted retinoid isomerase activity as well as prevented the characteristic loss of cone photoreceptors exhibited by this model.18 The rd12 mouse, a natural occurring RPE65 mutant, has also been extensively utilized as a model for gene therapy.19 AAV5-RPE65 improved ERG responses and visual guided behavior following subretinal injection in this strain.20 Later experiments, in which rd12 mice received subretinal injections of clinical grade AAV2-RPE65 aimed to establish an in vivo bioassay for characterizing AAV vector activity.21 In this study, delivery of vector spanning 2 log units, 10e8 to 10e10 vg/µl, resulted in a clear dose-response relationship.21 This bioassay was eventually used to evaluate stability of vector used during human clinical trials of LCA2.22,23
Due to their importance in daylight vision, particular attention has been focused on the fate of cone photoreceptors following AAV-RPE65 treatment. Cones utilize a noncanonical pathway for recycling of chromophore that is independent of the RPE, and enables them to function in continuous bright light.24,25 Studies of affected patients with LCA2 suggest that, on the whole, cones are still dependent on RPE65 isomerase activity for their survival, even though some residual cone activity persists in the absence of the isomerase.26 This is consistent with the natural histories of the Rpe65−/− and rd12 mice; cone photoreceptors degenerate relatively quickly.15,19 Several experiments in mouse models have specifically addressed cone survival as a consequence of gene therapy. Lentiviral and AAV-based vectors were used to treat RPE65 deficient mice that in some cases also lacked confounding rod function resulted in cone survival.27,28 Later it would be shown that self-complementary AAV-RPE65 vectors were capable of restoring cone function and preventing cone degeneration in both a rod “functionless ” RPE65 double knock-out mouse (Rho−/−Rpe65−/−) and the rd12 mouse.29
In several of the previously mentioned studies, investigators evaluated the ability of gene therapy to improve function in older affected animals, thereby modeling treatment in mid to late stage disease. In dog studies, measurable improvements in ERG and behavioral performance were noted for dogs treated as old as 30 months of age.30 Consistent visual improvements were possible in rd12 mice treated at 3 months of age.31 Even a small percentage (16%) of Rpe65−/− mice treated at 24 months of age showed significant improvements in ERG.17 Taken together, these results suggested that patients with LCA2 qualifying for phase I trials (i.e., as late as the 2nd decade of life or later), would have a reasonable chance of improved visual function upon treatment.
These proof of concept studies as well as Good Laboratory Practice safety studies would culminate with the multiple phase I/II gene therapy trials in human.22,32,33,34 Results of four clinical trials of RPE65 gene therapy for LCA2 have been independently initiated and interim outcomes published (NCT00481546, NCT00516477, NCT00643747; clinicaltrials.gov).22,23,32,33,34,35,36,37,38,39 The longest follow-up to date is 3 years.39 All three employed an AAV2 vector carrying a normal human RPE65 cDNA delivered subretinally to the worse eye. In spite of differences in the promoter employed, vector manufacturing details, dose and volumes delivered, anesthesia during vector delivery and postoperative steroid use, it was concluded by all studies that AAV-mediated subretinal gene therapy elicits no vector-related adverse events or toxic immune responses. All trials also reported clinical measures of vision improvement, but to various levels of detail and significance. A recent review of all reported clinical follow-up data up to 2010 has recently appeared.5 Since then, one more complete 3-year follow-up study was reported by Jacobson et al.,39 and the results of this study will be the primary focus here as it follows the largest cohort of treated patients for the longest time employing the most diverse measures of visual outcome.
Methods for measuring improvement in both light sensitivity and retinal function upon treatment of LCA2 had been previously developed in both mice and dog models of LCA2.8,21,40,41,42 When these methods were adapted to the clinical trials, most treated subjects reported qualitative improvements in dim light sensitivity within a short time after vector administration.22,32,34,35,36,37 To quantify these impressions, light sensitivity was quantified using a psychophysical full-field sensitivity threshold test (FST) that avoided the vision fixation instability common in these patients.14,42,43 In the 3-year study,39 there were statistically significant improvements from baseline in all 15 patients with posttreatment times varying among patients from 30 days to 3 years. FST improvements ranged from tenfold to 10,000-fold. The areas of dark-adapted (rod-mediated) visual field improvement, when assessed by microperimetry, corresponded well with the topological location of the subretinal bleb of vector mapped at the time of surgery, demonstrating that the retinal area responding to therapy is limited to that area receiving subretinal vector. Within this treated region, patients recovered as much as a 63,000 fold increase in their ability to detect in light. For some, this improvement was essentially all the functional gain possible given their baseline loss of photoreceptors.22,35 In patients with the highest light sensitivity, gains in both rod and cone function could be clearly discerned. In a test aimed at assessing the usefulness of this improved retinal function, the ability of treated subjects to navigate a walking maze was quantified with improvement relative to baseline seen in five of six patients.39
Visual acuity, classically quantified by the smallest size of high contrast letters readable at a fixed distance,44 was improved but with mixed statistical significance in nearly all patients in the three reported trials. However, in the context of retinal disease, particularly if foveal cones are affected, the location of highest acuity may not be within the normal human fovea.45 When vector was delivered subfoveally, necessarily accompanied by transient foveal detachment, patients with LCA2 often experienced no change in foveal light sensitivity but rather showed improvement extrafoveally.22,33,35 In the 3-year study,39 approximately half the patients experiencing a vector bleb that detached the fovea during surgery lost foveal thickness, presumably due to foveal cone loss, as assessed with follow-up optical coherence tomography (OCT) analysis. This suggests that foveal cones may be particularly sensitive to the potential damaging effects of subretinal vector-mediated foveal detachment, and that this locale for vector delivery should be cautiously approached in the future.
Related to foveal versus extrafoveal function after treatment is analysis of visual fixation, i.e., the retinal location used by a patient when asked to fix their gaze on an object they could see. At baseline, most patients in the 3-year study fixated in an area encompassing their fovea.39 Interestingly, at the 1-year follow-up visit but not before, one of the earliest patients treated reported the newly emergent ability to read the digital clock numbers in the family car.36 In an attempt to understand this apparently new visual perception at 1 year after treatment, the patient's fixation was determined in response to a dim target that could not be perceived at baseline or at any earlier treatment follow-up visit. The treated eye now exhibited a shift in fixation away from the fovea and solidly into the superior-temporal retina that coincided precisely with the locale of the original vector bleb. This phenomenon has been described previously under quite different circumstances: as a result of macular degeneration, loss of foveal cone function can lead to a gradual fixation shift into an extrafoveal location with better function than the anatomical fovea.46 This adaptation involves a change in central control of the ocular muscles to position images on this new, more light sensitive retinal region, the “pseudo-fovea”, and likely originates as a slow visual cortex response to a new and better functioning retinal locus47 arising through focal gene therapy. Presently, 6 of the 15 patients with LCA2 followed have developed similar pseudo-foveas upon multi-month delays after treatment (S. Jacobson and A. Cideciyan, personal communication, 2011). Such dramatic but relatively slow cortical plasticity in adults bodes well for other gene therapies for blinding retinal diseases that improve retinal function locally and suggests that the rate of generation of a useful pseudo-fovea may benefit from visual training.48
Most recently, readministration of AAV-RPE65 to the second, untreated contralateral eye is being investigated, with results out to 6 months indicating readministration is safe and efficacious in the previously naive eye.49
In summary, this first in human gene therapy for the RPE65 form of early retinal disease has been shown to be safe, free of serious complications, and effective at multiple clinical and visually useful levels. Efforts are currently underway to initiate a phase III clinical trial of AAV-RPE65 (NCT00999609; clinicaltrials.gov).
MERTK-associated autosomal recessive retinitis pigmentosa
The mer receptor tyrosine kinase (MERTK) is a member of the Axl/Mer/Tyro3 receptor tyrosine kinase family. MERTK is required for phagocytosis of photoreceptor outer segments by the RPE, and when absent leads to profound degeneration of the retina. The MERTK-associated form of arRP is very rare, with isolated patient populations identified in the Middle East and most recently the Faroe islands.50,51,52 The Royal College of Surgeons rat, first described in 1938 by Bourne and colleagues, is a widely used model of recessively inherited retinal degeneration.53,54 Retinal degeneration arises from the inability of the RPE to phagocytose shed PR outer segments, resulting in a subretinal debris field and subsequent photoreceptor loss via apoptosis.55,56,57 Degeneration of the retina is accompanied by progressive attenuation of retinal function as evaluated by ERG.58 In 2000, the retinal dystrophy locus of the Royal College of Surgeons rat was determined to be the MERTK gene.59 Follow-up studies identified patients with autosomal dominant retinitis pigmentosa (ADRP) with homozygous mutations in MERTK, conclusively linking this gene to the disease.60 Rescue experiments in the Royal College of Surgeons rat have included gene replacement as well as supplementation with neurotrophic factors aimed at arresting the degeneration and/or preserving the retina. In gene-replacement studies, adenovirus, AAV, and lentivirus have all been utilized.61,62,63 The most successful of these early studies utilized a lentivirus expressing MERTK and was able to preserve measurable retinal function out to 7 months postinjection.63 Interestingly, coadministration of lenti-Mertk and AAV expressing Glial-cell derived neurotrophic factor was more effective than lenti-MERTK alone.64 Most recently, an AAV8 Y-F capsid mutant vector was shown to preserve retinal structure and function in the Royal College of Surgeons rat out to at least 1 year after treatment.65 Presumably, the fast-acting nature of the AAV8 Y-F vector and relatively early (postnatal day 2) treatment restored MERTK expression before appreciable formation of a debris field, thus bypassing the need for supplementation with neuroprotective agents to prevent the downstream apoptosis of photoreceptors.65 A phase I clinical trial utilizing an AAV2 vector with an RPE-specific promoter driving MERTK has been initiated in Saudi Arabia using vector manufactured at the University of Florida (NCT014822195; clinicaltrials.gov). At this point in time, three patients have been treated by subretinal injection with no adverse events recorded (F. Alkuraya and E. Abboud, personal communication, 2012).
Usher syndrome
Usher syndrome (USH) refers to a clinically and genetically heterogeneous group of autosomal recessive disorders which account for the most frequent cause of combined deafness and blindness in humans with an estimated prevalence of 3–6 per 100,000 individuals, although that is likely an underestimation given the frequent false diagnosis of retinitis pigmentosa (RP) in infants.66,67 There are three clinical subtypes of USH (USH1, USH2, and USH3), distinguished by the severity and progression of hearing loss and the presence or absence of vestibular dysfunction, with visual loss due to RP being common to all three subtypes.68,69 USH1 is the most severe form in terms of the onset/extent of hearing loss and RP. Currently, mutations in five genes are known to be associated with USH1.70 The proteins encoded by these genes are expressed in cochlear hair cells of the inner ear as well as photoreceptors, both of which are highly specialized neurosensory cells with many structural and functional similarities. All five proteins are essential for the development and stability of the hair bundle in the inner ear as lack of their expression can prevent hair cell development altogether. Restoration of hearing via gene therapy may therefore only be possible in patients with milder USH where some hair cells are retained. In contrast, photoreceptors tend to develop normally but are progressively lost due to impaired protein transport through the connecting cilium and/or synapse dysfunction. Taken together, this suggests that in terms of gene replacement, a therapeutic window may exist primarily for the retinal phenotype of various forms of USH.
The first USH1 gene identified was myosinVIIa71 (MYO7A) which encodes an actin-based molecular motor that performs critical functions in both the inner ear and retina. Mutations in MYO7A cause Usher syndrome type 1b (Ush1b) and account for ~60% of all USH1.68 Patients with Ush1b are born profoundly deaf, have vestibular dysfunction and develop retinal degeneration in childhood.72 In the retina, MYO7A is expressed in RPE, the photoreceptor connecting cilia and photoreceptor synapses73,74,75 and plays a role in multiple cellular processes including intracellular transport, endocytosis and cell–cell adhesion.76 The shaker1 mouse model of Ush1b carries a mutation in Myo7a, is deaf and has vestibular dysfunction. Unlike patients with Ush1b, photoreceptors in this mouse model do not degenerate. A recent study demonstrates the existence of MYO7A in the calyceal processes of human photoreceptors and suggests that defects in this structure resulting from mutations in USH1 proteins are responsible for the retinal degeneration in patients with USH1.70 The lack of retinal degeneration in shaker1 mice (and other mouse models of USH) could be due to the fact that mice lack calyceal processes. However, shaker1 mice do have retinal phenotypes which distinguish them from wild type including mislocalized RPE melanosomes with defective motility and abnormal opsin transport through the connecting cilium.77,78,79,80 The requirement of MYO7A for the apical localization of melanosomes has also been demonstrated in human RPE cells.81
Initial proof-of-concept studies demonstrated that RPE melanosome localization and opsin transport could be restored in the shaker1 mouse following subretinal injection of a lentivirus carrying of Myo7a.82 These results led to a phase-1 clinical trial initiated by Oxford Biomedica UK employing an equine infectious anemia virus (EIAV) lentiviral vector to evaluate safety of subretinally delivered MYO7A in patients with Ush1b (NCT01505062; clinicaltrials.gov). However, lentiviral transduction is restricted primarily to the RPE following subretinal injection of postnatal retina.83,84 Because photoreceptors are the site of earliest disease expression,85 there is an obvious need to adequately transduce this cell type in patients with Ush1b. Following the assertion that AAV vectors were capable of packaging full-length MYO7A86 (~9 kb), work began to develop an AAV-based therapy for Ush1b. It is now understood that the packaging of large DNAs such as MYO7A initiates primarily from the 3′ end and proceeds until the AAV capsid reaches capacity (~5 kb). Resultant single-stranded vector DNAs are therefore heterogeneous in length and truncated at their 5′ ends.87,88,89 It is thought that full-length large-gene reconstitution occurs from these incomplete cDNAs via either recombination between overlapping homologous internal cDNA sequences or direct annealing of vector-packaged, opposite-polarity large-transgene fragments followed by host cell DNA repair synthesis on both DNA strands primed by their free 3′ ends.87,88,89 Such “heterogeneous ” AAV vector technology has since been used to deliver Myo7a to the subretinal space of shaker1 mice. Both serotype 2 and serotype 5 AAV vectors were capable of delivering full-length Myo7a to photoreceptors and RPE and correcting both melanosome localization and opsin transport in treated shaker1 mice.90,91 A hurdle for clinical application of heterogeneous vectors is the inability to discretely characterize the genomes within each capsid. As such, development of dual AAV vector platforms are currently underway for the treatment of Ush1b using vectors containing discretely characterized DNA payloads (Supplementary Figure S2). These will be described in more detail in the online supplement.
Stargardt disease, recessive ABCA4 form
Stargardt disease is a common form of juvenile macular degeneration, the most prevalent form being recessively inherited and associated with mutations in the photoreceptor-specific ABCA4, a member of the superfamily of ATP-binding cassette (ABC) transporters.92 ABCA4 functions as a “flippase ” of N-retinylidene-phosphatidylethanolamine, thereby transporting it from the lumen to the cytoplasmic side of photoreceptor outer disks.93 Mutations in ABCA4 result in decreased transport of N-retinylidene-phosphatidylethanolamine and phosphatidylethanolamine which is thought to lead to the formation of toxic retinoid compounds, eventually manifesting as an accumulation of lipid known as lipofuscin in the RPE, primarily composed of N-retinylidene-N-retinylethanolamine (A2E).94 The structure/function and biology of ABCA4 have been recently reviewed by Molday et al. (insert same reference as above). Mice lacking abca4 (abca4−/− or alternatively abcr−/−), the murine homolog to ABCA4, also show an increased accumulation of A2E and the formation of lipofusin granules.95 The size of the ABCA4 cDNA is 6.8 KB and therefore beyond the conventional packaging capacity of AAV. As such, the same approaches for gene replacement discussed for MYO7A Ushers1b (“heterogeneous ” AAV and EIAV lentivirus) have been employed in the abca4−/− mouse.86,96 In both studies, correction of phenotype (improved recovery of photoreceptor desensitization and/or reduction of liposfuscin) in treated abca4−/− was observed. It is important to note that in the EIAV lentivirus study, injections were performed at P4 to P5, before photoreceptors had reached terminal differentiation, whereas AAV injections were performed at 1 month of age, in postmitotic retina.86,96 On the basis of the positive results reported in Kong et al.,96 Oxford BioMedica has initiated a phase I/II clinical trial utilizing the EIAV lentiviral platform to deliver ABCA4 to patients with Stargardt (NCT01367444; clinicaltrials.gov). Most recently, nanoparticle delivery of ABCA497 into abca4−/− mice has also show to improve the retinal phenotype using the same criteria as mentioned above.
Choroideremia
Mutations in the CHM gene are associated with choroideremia, an X-linked retinal degeneration affecting 1:50,000 males that is readily diagnosed with fundus examination.98,99,100,101,102 Initially, patients exhibit retinal thickening, followed by photoreceptor degeneration, RPE depigmentation, and retinal remodeling. Affected children and carrier females have mottled areas of pigmentation whereas affected males display loss of RPE and choriocapillaris. Affected males develop night blindness in their teens which progresses to loss of peripheral visual field and more profound blindness within three decades of onset.103,104
CHM encodes Rab Escort Protein 1 (REP-1), a ubiquitously expressed protein required for the efficient geranylgeranylation of ras-related GTPases, or Rab proteins, which are integral to the trafficking of vesicles in endocytic and exocytic pathways.100,105,106 Geranylgeranylation, or prenylation, requires the activity of both a transferase (RabGGTase) and REP-1 which tethers the unprenylated Rab substrate to the RabGGTase and then “escorts ” the newly prenylated Rab protein to a specific cellular membrane.107 Patients with chroroideremia express little or no REP-1,98,108,109 and it has been suggested that this lack of protein affects opsin transport to photoreceptor outer segments, apical migration of RPE melanosomes and the phagocytosis of photoreceptor outer segments by the RPE.110 Indeed, siRNA knockdown of REP-1 in human fetal RPE cells revealed that photoreceptor outer segment internalization was unaffected but that protein clearance was delayed due to inhibition of phagosome–lysosome fusion events.111 Generation of conditional knock-out mice revealed that disease pathogenesis involves independently triggered degeneration of photoreceptors and RPE, each associated with their own Rab prenylation defects.112 Newer conditional mutants in which Chm is knocked out in either the RPE or photoreceptors, independently, reveal that despite the cell autonomous hallmarks of this disease, RPE defects accelerate photoreceptor degeneration.113
As a first step towards developing a clinical gene therapy trial for choroideremia, recombinant adenovirus was used to deliver full-length human REP-1 to defective lymphocytes and fibroblasts isolated from patients with choroideremia.114 Investigators showed that vector-mediated REP-1 expression restored RabGGTase activity in vitro. More recently, lentiviral-mediated delivery of REP-1 to the RPE-specific Chm knock-out mouse revealed increases in prenylation activity in the RPE of treated mice.115 Although this study served as proof of concept for restoring function to cells carrying Chm mutations, the authors acknowledged that more efficient photoreceptor transduction might be required to restore function to the neural retina. In the fall of 2011, a clinical trial for the treatment of choroideremia was initiated. This 12-subject safety study employed an AAV2 vector to deliver REP-1 to the subretinal space of affected patients.116 Treatment safety and efficacy will be evaluated over the next 1.5 years.
Exudative (wet) age-related macular degeneration
Vascular endothelial growth factor (VEGF) is the major proangiogenic factor promoting neovascularization of the choroidal vasculature in exudative (wet) age-related macular degeneration (AMD), the leading cause of blindness in those above 65.117,118 Inhibition of VEGF with antibodies, RNA aptamers or soluble receptors has been clinically tested for managing AMD and has shown promise.119,120,121,122 Current practice using various versions of VEGF antibody requires long term and repeated intravitreal injections and invariably involves the cumulative risk of repeated intraocular injections and significant financial burden. Soluble FLT-1 (sFLT1) is a portion of the R1 VEGF receptor that binds extracellular VEGF and prevents it from binding to its normal cell surface vascular endothelial receptors FLT-1 and FLK. Ocular neovascularization was inhibited in animal models upon subretinal AAV2 carrying full-length sFlt1 (domains 1–7)123,124 and treatment was also shown to be safe in nonhuman primates.125
Recently, an AAV2 vector with a novel soluble chimeric protein (AAV2-sFLT01) was shown to be both persistently expressed after a single injection and therapeutic at controlling neovascularization in a murine model when delivered intravitreally,126 a much less traumatic route of administration than subretinal vector. The protein encoded by this vector is a hybrid comprised of Flt-1 domain 2 linked to a human immunoglobulin G1 heavy chain Fc fragment. When expressed, an extracellular homodimer forms that binds to VEGF with high affinity and avidity. Intravitreal AAV2-sFLT01 efficacy was confirmed in two mouse models of ocular neovascularization, the oxygen-induced retinopathy of prematurity model and the laser-induced choroidal neovascularization (CNV) model127,128 and was recently extended to an AAV8 vector.129 The Lukason et al. study128 also included analysis of therapy in the nonhuman primate laser-induced CNV model of wet AMD. Stable sFLT01 expression was observed for 5 months in the aqueous humor. Laser treatment was initiated at 22 weeks postvector injection, and a significant reduction of CNV was documented in seven of eight treated eyes whose levels of sFLT01 in the aqueous humor exceeded 100 ng/ml, suggesting a potential minimum effective level of sFLT01 is needed to observe a reduction in CNV. Based on these data and formal safety/biodistribution studies establishing safe vector doses in the monkey, Genzyme/Sanofi has initiated a phase-1 clinical trial using intravitreal AAV2-sFLT01 for neovascular AMD130 (NCT01024998; clinicaltrials.gov).
Endostatin, a collagen XVIII cleavage product, and angiostatin, a fibrinogen cleavage product, both inhibit tumor angiogenesis.131,132 With adenoviral 5-endostatin vectors delivered systemically to laser CNV mice, serum endostatin levels correlated well with CNV inhibition.133 More direct vitreal injection of either AAV-endostatin,134 AAV-angiostatin (K1-3 domains)135 or lentiviral vector-angiostatin136 also significantly inhibited murine CNV. These data have led in part to a phase-1 clinical trial initiated by Oxford Biomedica (Oxford, UK) employing an EIAV lentiviral vector expressing both endostatin and angiostatin for late stage AMD (NCT01301443; clinicaltrials.gov). Results are pending.
PreclinIcal
GUCY2D-Leber congenital amaurosis
One of the most frequently mutated LCA genes is guanylate cyclase-1 (GUCY2D).137,138,139 A recent study evaluating the spectrum of causative mutations in an Asian population of 87 patients with LCA identified mutations in GUCY2D as the most common cause of the disease (~16%).140 GUCY2D encodes the retina-specific protein guanylate cyclase-1 (GC1) which is expressed in the outer segments of rod and cone photoreceptors of human, monkey and mouse retinas.141,142 GC1 plays a vital role in the light–dark and recovery cycle, anchoring, via cGMP, the feedback loop linking intracellular calcium levels and the polarization state of photoreceptors.143 Like other membrane guanylatecyclases, it contains an N′-terminal signal sequence, an extracellular domain, a single transmembrane domain, a kinase-like homology domain, a dimerization domain and a C′-terminal catalytic domain, and is present likely as a homodimer.144 LCA1-causing mutations are distributed throughout GC1,145,146 can alter enzyme structure and stability, impact retrograde transport of other peripheral membrane associated proteins and are frequently null.145,146 Mutations that reduce or abolish the ability of GC1 to replenish intracellular cGMP and reopen cGMP-gated cation channels, as is the case in LCA1, are thought to create the biochemical equivalent of chronic light exposure in rod and cone photoreceptors. A hallmark feature of LCA1 is preserved retinal appearance. Patients present in infancy with a normal fundus and, despite having an unrecordable cone ERG and abnormal/unrecordable rod ERG, patients retain normal photoreceptor laminar architecture except for mainly foveal cone outer segment abnormalities.138,146,147,148 Maintenance of retinal structure in LCA1 is unlike other forms of the LCA which exhibit marked retinal thinning that generally worsens with age. Taken together, the prevalence of GUCY2D mutations and the maintenance of retinal structure in patients with LCA1 over time suggest they may be good candidates for gene-replacement therapy.
The GC1 knock-out (GC1ko) mouse exhibits a loss of cone function that precedes cone degeneration.149 Rod photoreceptors maintain 30–50% of their function and do not degenerate, a result likely due to the presence of guanylate cyclase-2 (GC2), a close relative of GC1, in those cells. AAV5 vectors containing Gucy2e (the mouse homologue of GUCY2D) and either the ubiquitous smCBA or photoreceptor-specific human rhodopsin kinase (hGRK1) promoter restored cone function (ERG), cone-mediated behavior and preserved cone photoreceptors in the GC1KO mouse for at least 3 months.150 Subsequent studies have demonstrated that AAV8 and AAV5 based vectors containing the hGRK1 promoter driving either GUCY2D or Gucy2e, respectively, restore photoreceptor function, cone-mediated behavior and preserved cones over the long term.151,152 These results suggest that therapy persists over the lifetime of the treated animal. Because patients with LCA1 have either absent or abnormal rod photoreceptor function, it was important to ask whether AAV-mediated GC1 expression could restore function to rods in a model with physiologically “silent ” photoreceptors. The GC1/GC2 double knock-out (GCdko) mouse was generated153 to suit this need. Like some patients with LCA1, these mice also exhibit complete loss of both rod and cone function. AAV5 and AAV8 (Y733F) vectors containing the hGRK1 promoter and murine GC1 proved capable of restoring both rod and cone function (ERG), rod- and cone-mediated visual behavior and preserving rod and cone structure following subretinal delivery to the GCdko mouse.154 Related to this, a recent study showed that the hGRK1 promoter has exclusive activity in both rods and cones following subretinal injection of AAV5-hGRK1-GFP in nonhuman primate.155 This study validated that AAV5 is capable of transducing foveal, parafoveal, and perifoveal cones as well as rods of NHP (Supplementary Table S1). Taken together, these results suggest that AAV5 containing the hGRK1 promoter and GUCY2D cDNA is a suitable choice of clinical vector intended for the treatment of LCA1.
Achromatopsia
The complete form of achromatopsia (ACHM) is an autosomal recessive disease that affects approximately 1 in 30,000 individuals and is associated with the loss or absence of cone function. ACHM is marked by poor central visual acuity (<20/200) as well as photophobia, complete color blindness and reduced cone-mediated ERG response amplitudes. It is an attractive disease for gene-replacement therapy as cone photoreceptors are present and often fairly intact in affected individuals. Although initially thought to be a stationary disease based on normal macular appearance, it has recently been shown that cone degeneration begins early in childhood, with degeneration progressing at a modest rate.156 Mutations in genes encoding subunits of the cone-specification channel, cyclic nucleotide gated channel α and β 3, CNGA3 and CNGB3, account for a combined 80% of all ACHM cases.157,158 Mutations in two genes associated with phototransduction in cones, cone-specific α subunit of transducin, GNAT2, and α subunit of cone-specific phosphodiesterase, PDE6C are much rarer, accounting for perhaps no more than 5% of cases combined.159,160,161 The first report of gene therapy for ACHM was performed in the Gnat2cpfl3 mouse162 which carries a recessive mutation in Gnat2 resulting in little to no cone-mediated ERG and poor visual acuity.162,163 Subretinal injection of AAV5 driving Gnat2 under the control of the human red cone opsin promoter, restored cone mediated ERG amplitudes and cone-mediated behavioral responses to levels indistinguishable from age-matched wild-type mice.162 Gene-replacement therapy has also been tested in two large animal models of ACHM. AAV5 containing human CNGB3 was used to treat two independent canine models of CNGB3-ACHM.164 Vector-treated dogs exhibited restored cone function and day vision, however the magnitude and persistence of therapy was dependent on both the age at treatment and the promoter used.164 Affected dogs that were treated at a relatively young age with vector utilizing the human red cone opsin promoter exhibited stable treatment effect out to at least 33 months.164 Importantly, GNAT2 and CNGA3 which are absent or mislocalized in untreated cones, showed normalization of protein expression and localization to cone outer segments in treated eyes.164 Gene-replacement studies in mouse models of both the CNGA3 and CNGB3 forms of ACHM have also shown success using AAV5 and AAV8 based vectors.165,166,167 As with the CNGB3 dog study, treated mouse eyes show a restoration of expression and normal localization of cone-specific proteins following treatment.165,166,167 Given the high density of cones in the macula and the previously mentioned challenges encountered when delivering vector subretinally under the macula, the development of vectors capable of transducing photoreceptors following intravitreal delivery will be highly advantageous for moving ACHM gene-replacement therapy into the clinical trial phase.
X-linked juvenile retinoschisis
X-linked juvenile retinoschisis (XLRS) is the leading cause of monogenic macular dystrophy in males with a prevalence between 1:5,000 and 1:25,000.168 It is characterized by localized schisis, or splitting in the retina which can occur in all layers.169,170 Patients with XLRS exhibit an unusual electronegative ERG wherein the a-wave is relatively preserved whereas the b-wave is severely diminished171 due presumably to a relatively preserved photoreceptor response but a loss of interaction with second order retinal neurons. XLRS is considered a stationary condition with retinal presentation in early childhood, exacerbation of symptoms in teenage years and stabilization in adulthood.168,172 Schisis lesions become less obvious in the fourth to fifth decade but complications may still arise from macular atrophy, vitreous hemorrhage, or retinal detachment.
XLRS is caused by mutations in the retinoschisin (RS1) gene which encodes a 24 kDa protein (RS1) that is expressed in the retina and pineal gland.173,174 RS1 is secreted from retinal neurons, most likely photoreceptors, as a homo-octamer that adheres to retinal cell surfaces.175 Although the function of this protein is not completely understood, its discoidin domain suggests that, like other discoidin domain-containing proteins, it may be involved in cell adhesion or cell-extracellular matrix interactions.175,176 The RS1 knock-out (Rs1h-KO) mouse yields many hallmark features of the human condition including electronegative ERG waveforms and the presence of schisis cavities.177,178,179 Several groups have reported that gene supplementation of the normal Rs1h protein in Rs1h-KO mice confers therapy to this mouse model. Initially, it was shown that intravitreal delivery at 13 weeks of age of a serotype 2 AAV vector containing the ubiquitous cytomegalovirus promoter and the murine RS1 cDNA to Rs1h-KO mice restored function (ERG) to this model out to 6 months of age.177 Subretinal injections of serotype 5 AAV containing the photoreceptor-specific murine opsin promoter and the human RS1 cDNA into younger Rs1h-KO mice (P14) revealed more pronounced preservation of retinal structure and function.180 Using the initial AAV2 vector, long-term (14 month) functional and structural improvements were next reported following P14, intravitreal injection.179 Window of therapy was addressed in a subsequent report showing that AAV5-murine opsin-hRS1 vector improved retinal structure and function following subretinal injections in P15, 1 month and 2 month old Rs1h-KO mice (Supplementary Figure S1). Injections at 7 months of age, however, improved only retinal structure (assayed via photoreceptor survival and retention of cone outer segments) and not ERG.181 A closer examination of protein expression and morphology of the outer plexiform layer revealed that P14 injection of AAV2-cytomegalovirus-mRS1 resulted in increases in postsynaptic density protein (PSD95) and bipolar cell metabotropic glutamate receptor subtype 5 (mGluR6), decreases in glial fibrillary acidic protein and improvements in retinal function out to 8 months of age.182 Most recently, it was reported that a serotype 8 AAV vector containing a truncated human retinoschisin promoter (hRSp4) and the human RS1 cDNA was capable of improving structure and function out to 15 weeks after intravitreal delivery to 6–7 week old Rs1h-KO mice.183 These studies highlight that multiple parameters including delivery method, vector serotype and promoter should be carefully considered prior to clinical application of an XLRS gene therapy vector.
Autosomal dominant RP
Mutations in at least 22 genes lead to ADRP. Diseases may be dominantly inherited for three reasons: haploinsufficiency, dominant negative gene product or toxic gain-of-function. Haploinsufficiency implies that the amount of product produced by a single wild-type allele does not support normal function. Dominant negative mutations typically interfere with the ability of the wild-type protein to reach its appropriate destination in the cell or to assemble in a multi-subunit complex. Mutations in ELOVL4 appear to be dominant for this reason.184 Toxic gain-of-function mutations create proteins that are themselves harmful to the cell. An example in the retina would be the Q344ter mutation in rhodopsin (RHO), which causes rhodopsin to mislocalize to the cell bodies of rod photoreceptor cells.185
In the case of haploinsufficiency, gene therapy requires delivery of a wild-type cDNA so that the level of the normal protein becomes sufficient to support retinal health. Cai et al. have used compacted DNA nanoparticles to treat a dominant loss-of-function of mutation of PRPH2 in mouse model of retinal degeneration.186 Gene therapy for dominant negative mutations or for toxic mutations is more complicated, however, since the dominant allele may require silencing. Several groups have employed small RNA technology including ribozymes and RNA interference to suppress the production of mutant. For example, in a rat model of ADRP caused by the P23H mutation of rhodopsin, AAV delivery of allele specific hairpin and hammerhead ribozymes led to long-term (6 month) rescue of photoreceptors as measured by histology and by the ERG response.187,188 The major obstacle to mutation-specific silencing is allelic heterogeneity. There are more than 100 dominant mutations in the RHO gene alone, and it is impractical to generate a sequence specific inhibitor for each mutation. For this reason, a mutation-independent approach in which ribozymes, or more recently siRNAs, are being designed to target regions of mRNA that are not affected by mutation, so that both wild-type and mutant RNA are degraded.189 As the RNA inhibitors (ribozymes, siRNAs or synthetic miRNAs) themselves lead to retinal degeneration, for therapy, they must be coupled with the delivery and expression of cDNAs bearing silent mutations that render them resistant to the codelivered ribozyme or siRNA.190,191,192,193,194,195,196,197 Millington-Ward et al. recently used coinjection of 2 AAV vectors to significantly reduce the rate of retinal degeneration in a mouse line bearing a dominant P347S mutant transgene.194 Mao et al. demonstrated that delivery of an siRNA designed to target mouse Rho and human P23H RHO mRNA as well as a resistant RHO gene, both contained within a single AAV vector, conferred long-term (9 month) rescue of retinal function and preservation of retinal structure following subretinal injection of P23H mice.198
Instead of inhibiting gene expression by degrading mutant mRNAs, Mussolino and colleagues used zinc-finger transcription factors to suppress the synthesis of P347S rhodopsin transgenic mouse model of autosomal dominant RP.199 Silencing of the mutant human transgene led to increased viability and function of photoreceptors in the region of the retina infected by AAV. By reducing the size of the promoter of the gene targeted by the transcription factor (e.g., human RHO), a resistant wild-type gene could be delivered in conjunction with the Zn-finger protein.
However the “suppress and supplant ” approach to gene therapy may not be necessary for treatment of dominant mutations in the RHO gene. In a mouse model of ADRP caused by a P23H human RHO transgene, Mao et al. recently demonstrated that simple AAV delivery of a wild-type mouse cDNA could significantly retard retinal degeneration as measured by ERG amplitudes and morphometry.200 They concluded that P23H must be a dominant negative rather than a toxic gain-of-function mutation, and that treatment could be effected by altering the balance between mutant and wild-type forms of rhodopsin.
Because protein misfolding is believed to contribute to the pathogenesis associated with a large class of rhodopsin mutations, gene transfer of molecular chaperones is also an attractive approach for ADRP therapy. Gorbatyuk and colleagues used AAV to increase the expression of Grp78, an Hsp70 family member that resides in the endoplasmic reticulum201 and reported significant improvement in ERG amplitudes and survival of photoreceptors in treated eyes. This rescue was not associated with increases in the level of properly reconstituted rhodopsin but rather with suppression of the unfolded protein response.
Leber hereditary optic neuropathy
Mitochondrial Leber hereditary optic neuropathy (LHON) is a retinal disease of ganglion cells that leads to blindness in young adults.202 A G-to-A transition in mitochondrial DNA at position 11778 in the gene encoding subunit 4 of NADH dehydrogenase (ND4) leads to a position 340 arginine-to-histidine substitution that is responsible for approximately half of LHON cases. In contrast to other mitochondrial diseases, LHON vision loss requires essentially all copies of the mitochondrial DNA to be mutated. Therefore, a small amount of normally functioning ND4 is likely to be therapeutic and may protect against blindness. Until recently,203 there had been no reports of successfully delivering a DNA to mammalian mitochondria, so this approach had not been an option. One alternative for ND4 LHON is to express wild-type ND4 protein in the cytoplasm by delivering an ND4 cDNA to the nucleus that is recoded in the cytoplasmic triplet code rather than the mitochondrial code and, in addition, contains an appended N-terminal mitochondrial targeting sequence to mediate efficient organelle import as has been done earlier in yeast.204,205 Qi et al.206 showed that this approach would create the LHON phenotype in a normal mouse by delivering mutant 11778 human ND4 cDNA in an AAV 2 vector to the mitochondria of retinal ganglion cells (RGCs) in a normal mouse. Using electrophoresis to transduce RGCs, Ellouze et al.207 then similarly introduced the mutant 11778 human ND4 into rats and showed that mitochondrially targeted allotopic wild-type ND4 was sufficient for rescue of RGC loss and visual function induced earlier by delivery of mutant G11778A ND4. The safety of such an allotopic wild-type human ND4 gene delivery was then shown in a mouse with a FLAG-tagged, recoded, mitochondrially retargeted human ND4 delivered intravitreally by an AAV2 vector with the CBA promoter.208 FLAG-tagged ND4 transgene product was localized to the mitochondria of RGCs. Moreover, treatment did not result in a loss of RGCs, alteration in the rates of mitochondrial ATP synthesis or loss in pattern evoked ERG signals. Together, these data support the idea that allotopic mitochondrial gene delivery can be an effective and safe mode of therapy for LHON.
SUPPLEMENTARY MATERIAL Table S1. Transduction profiles of AAV serotypes across species. Figure S1. Schematic of subretinal versus intravitreal injection routes. Figure S2. Dual AAV vector platforms. Data.
Acknowledgments
We thank Jim Peterson for his editorial assistance. We acknowledge NIH grants EY021721, R24EY022012, T32 EY00713 and grants from the Macula Vision Research Foundation, Foundation Fighting Blindness, Eldon Family Foundation, Creed's Cause Foundation, Vision for Children and Research to Prevent Blindness, Inc. for partial support of this work. W.W.H. and the University of Florida have a financial interest in the use of AAV therapies, and own equity in a company (AGTC Inc.) that might, in the future, commercialize some aspects of this work. The other authors declare no conflicts of interest.
Supplementary Material
References
- Rodieck RW. Sinauer Associates: Sunderland; 1998. The First Steps in Seeing. [Google Scholar]
- Jin M, Li S, Moghrabi WN, Sun H., and, Travis GH. Rpe65 is the retinoid isomerase in bovine retinal pigment epithelium. Cell. 2005;122:449–459. doi: 10.1016/j.cell.2005.06.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moiseyev G, Chen Y, Takahashi Y, Wu BX., and, Ma JX. RPE65 is the isomerohydrolase in the retinoid visual cycle. Proc Natl Acad Sci USA. 2005;102:12413–12418. doi: 10.1073/pnas.0503460102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Redmond TM, Poliakov E, Yu S, Tsai JY, Lu Z., and, Gentleman S. Mutation of key residues of RPE65 abolishes its enzymatic role as isomerohydrolase in the visual cycle. Proc Natl Acad Sci USA. 2005;102:13658–13663. doi: 10.1073/pnas.0504167102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV. Leber congenital amaurosis due to RPE65 mutations and its treatment with gene therapy. Prog Retin Eye Res. 2010;29:398–427. doi: 10.1016/j.preteyeres.2010.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Acland GM, Aguirre GD, Ray J, Zhang Q, Aleman TS, Cideciyan AV.et al. (2001Gene therapy restores vision in a canine model of childhood blindness Nat Genet 2892–95. [DOI] [PubMed] [Google Scholar]
- Narfström K, Katz ML, Bragadottir R, Seeliger M, Boulanger A, Redmond TM.et al. (2003Functional and structural recovery of the retina after gene therapy in the RPE65 null mutation dog Invest Ophthalmol Vis Sci 441663–1672. [DOI] [PubMed] [Google Scholar]
- Acland GM, Aguirre GD, Bennett J, Aleman TS, Cideciyan AV, Bennicelli J.et al. (2005Long-term restoration of rod and cone vision by single dose rAAV-mediated gene transfer to the retina in a canine model of childhood blindness Mol Ther 121072–1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Narfström K, Vaegan, Katz M, Bragadottir R, Rakoczy EP., and, Seeliger M. Assessment of structure and function over a 3-year period after gene transfer in RPE65-/- dogs. Doc Ophthalmol. 2005;111:39–48. doi: 10.1007/s10633-005-3159-0. [DOI] [PubMed] [Google Scholar]
- Jacobson SG, Acland GM, Aguirre GD, Aleman TS, Schwartz SB, Cideciyan AV.et al. (2006Safety of recombinant adeno-associated virus type 2-RPE65 vector delivered by ocular subretinal injection Mol Ther 131074–1084. [DOI] [PubMed] [Google Scholar]
- Le Meur G, Stieger K, Smith AJ, Weber M, Deschamps JY, Nivard D.et al. (2007Restoration of vision in RPE65-deficient Briard dogs using an AAV serotype 4 vector that specifically targets the retinal pigmented epithelium Gene Ther 14292–303. [DOI] [PubMed] [Google Scholar]
- Bennicelli J, Wright JF, Komaromy A, Jacobs JB, Hauck B, Zelenaia O.et al. (2008Reversal of blindness in animal models of leber congenital amaurosis using optimized AAV2-mediated gene transfer Mol Ther 16458–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Narfström K, Seeliger M, Lai CM, Vaegan, Katz M, Rakoczy EP.et al. (2008Morphological aspects related to long-term functional improvement of the retina in the 4 years following rAAV-mediated gene transfer in the RPE65 null mutation dog Adv Exp Med Biol 613139–146. [DOI] [PubMed] [Google Scholar]
- Aguirre GK, Komáromy AM, Cideciyan AV, Brainard DH, Aleman TS, Roman AJ.et al. (2007Canine and human visual cortex intact and responsive despite early retinal blindness from RPE65 mutation PLoS Med 4e230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Redmond TM, Yu S, Lee E, Bok D, Hamasaki D, Chen N.et al. (1998Rpe65 is necessary for production of 11-cis-vitamin A in the retinal visual cycle Nat Genet 20344–351. [DOI] [PubMed] [Google Scholar]
- Dejneka NS, Surace EM, Aleman TS, Cideciyan AV, Lyubarsky A, Savchenko A.et al. (2004In utero gene therapy rescues vision in a murine model of congenital blindness Mol Ther 9182–188. [DOI] [PubMed] [Google Scholar]
- Jacobson SG, Aleman TS, Cideciyan AV, Sumaroka A, Schwartz SB, Windsor EA.et al. (2005Identifying photoreceptors in blind eyes caused by RPE65 mutations: Prerequisite for human gene therapy success Proc Natl Acad Sci USA 1026177–6182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y, Moiseyev G, Takahashi Y., and, Ma JX. RPE65 gene delivery restores isomerohydrolase activity and prevents early cone loss in Rpe65-/- mice. Invest Ophthalmol Vis Sci. 2006;47:1177–1184. doi: 10.1167/iovs.05-0965. [DOI] [PubMed] [Google Scholar]
- Pang JJ, Chang B, Hawes NL, Hurd RE, Davisson MT, Li J.et al. (2005Retinal degeneration 12 (rd12): a new, spontaneously arising mouse model for human Leber congenital amaurosis (LCA) Mol Vis 11152–162. [PubMed] [Google Scholar]
- Pang JJ, Chang B, Kumar A, Nusinowitz S, Noorwez SM, Li J.et al. (2006Gene therapy restores vision-dependent behavior as well as retinal structure and function in a mouse model of RPE65 Leber congenital amaurosis Mol Ther 13565–572. [DOI] [PubMed] [Google Scholar]
- Roman AJ, Boye SL, Aleman TS, Pang JJ, McDowell JH, Boye SE.et al. (2007Electroretinographic analyses of Rpe65-mutant rd12 mice: developing an in vivo bioassay for human gene therapy trials of Leber congenital amaurosis Mol Vis 131701–1710. [PubMed] [Google Scholar]
- Cideciyan AV, Aleman TS, Boye SL, Schwartz SB, Kaushal S, Roman AJ.et al. (2008Human gene therapy for RPE65 isomerase deficiency activates the retinoid cycle of vision but with slow rod kinetics Proc Natl Acad Sci USA 10515112–15117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banin E, Bandah-Rozenfeld D, Obolensky A, Cideciyan AV, Aleman TS, Marks-Ohana D.et al. (2010Molecular anthropology meets genetic medicine to treat blindness in the North African Jewish population: human gene therapy initiated in Israel Hum Gene Ther 211749–1757. [DOI] [PubMed] [Google Scholar]
- Wang JS, Estevez ME, Cornwall MC., and, Kefalov VJ. Intra-retinal visual cycle required for rapid and complete cone dark adaptation. Nat Neurosci. 2009;12:295–302. doi: 10.1038/nn.2258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang JS., and, Kefalov VJ. An alternative pathway mediates the mouse and human cone visual cycle. Curr Biol. 2009;19:1665–1669. doi: 10.1016/j.cub.2009.07.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Aleman TS, Cideciyan AV, Heon E, Golczak M, Beltran WA.et al. (2007Human cone photoreceptor dependence on RPE65 isomerase Proc Natl Acad Sci USA 10415123–15128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bemelmans AP, Kostic C, Hornfeld D, Jaquet M, Crippa SV, Hauswirth WW.et al. (2006Lentiviral vectors containing a retinal pigment epithelium specific promoter for leber congenital amaurosis gene therapy. Lentiviral gene therapy for LCA Adv Exp Med Biol 572247–253. [DOI] [PubMed] [Google Scholar]
- Bemelmans AP, Kostic C, Crippa SV, Hauswirth WW, Lem J, Munier FL.et al. (2006Lentiviral gene transfer of RPE65 rescues survival and function of cones in a mouse model of Leber congenital amaurosis PLoS Med 3e347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pang J, Boye SE, Lei B, Boye SL, Everhart D, Ryals R.et al. (2010Self-complementary AAV-mediated gene therapy restores cone function and prevents cone degeneration in two models of Rpe65 deficiency Gene Ther 17815–826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Narfström K, Katz ML, Ford M, Redmond TM, Rakoczy E., and, Bragadóttir R. In vivo gene therapy in young and adult RPE65-/- dogs produces long-term visual improvement. J Hered. 2003;94:31–37. doi: 10.1093/jhered/esg015. [DOI] [PubMed] [Google Scholar]
- Li X, Li W, Dai X, Kong F, Zheng Q, Zhou X.et al. (2011Gene therapy rescues cone structure and function in the 3-month-old rd12 mouse: a model for midcourse RPE65 leber congenital amaurosis Invest Ophthalmol Vis Sci 527–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maguire AM, Simonelli F, Pierce EA, Pugh EN, Jr, Mingozzi F, Bennicelli J.et al. (2008Safety and efficacy of gene transfer for Leber's congenital amaurosis N Engl J Med 3582240–2248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bainbridge JW, Smith AJ, Barker SS, Robbie S, Henderson R, Balaggan K.et al. (2008Effect of gene therapy on visual function in Leber's congenital amaurosis N Engl J Med 3582231–2239. [DOI] [PubMed] [Google Scholar]
- Hauswirth WW, Aleman TS, Kaushal S, Cideciyan AV, Schwartz SB, Wang L.et al. (2008Treatment of leber congenital amaurosis due to RPE65 mutations by ocular subretinal injection of adeno-associated virus gene vector: short-term results of a phase I trial Hum Gene Ther 19979–990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Hauswirth WW, Aleman TS, Kaushal S, Schwartz SB, Boye SL.et al. (2009Human RPE65 gene therapy for Leber congenital amaurosis: persistence of early visual improvements and safety at 1 year Hum Gene Ther 20999–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Hauswirth WW, Aleman TS, Kaushal S, Schwartz SB, Boye SL.et al. (2009Vision 1 year after gene therapy for Leber's congenital amaurosis N Engl J Med 361725–727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maguire AM, High KA, Auricchio A, Wright JF, Pierce EA, Testa F.et al. (2009Age-dependent effects of RPE65 gene therapy for Leber's congenital amaurosis: a phase 1 dose-escalation trial Lancet 3741597–1605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simonelli F, Maguire AM, Testa F, Pierce EA, Mingozzi F, Bennicelli JL.et al. (2010Gene therapy for Leber's congenital amaurosis is safe and effective through 1.5 years after vector administration Mol Ther 18643–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Cideciyan AV, Ratnakaram R, Heon E, Schwartz SB, Roman AJ.et al. (2012Gene therapy for leber congenital amaurosis caused by RPE65 mutations: safety and efficacy in 15 children and adults followed up to 3 years Arch Ophthalmol 1309–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Hooser JP, Aleman TS, He YG, Cideciyan AV, Kuksa V, Pittler SJ.et al. (2000Rapid restoration of visual pigment and function with oral retinoid in a mouse model of childhood blindness Proc Natl Acad Sci USA 978623–8628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Hooser JP, Liang Y, Maeda T, Kuksa V, Jang GF, He YG.et al. (2002Recovery of visual functions in a mouse model of Leber congenital amaurosis J Biol Chem 27719173–19182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roman AJ, Cideciyan AV, Aleman TS., and, Jacobson SG. Full-field stimulus testing (FST) to quantify visual perception in severely blind candidates for treatment trials. Physiol Meas. 2007;28:N51–N56. doi: 10.1088/0967-3334/28/8/N02. [DOI] [PubMed] [Google Scholar]
- Roman AJ, Schwartz SB, Aleman TS, Cideciyan AV, Chico JD, Windsor EA.et al. (2005Quantifying rod photoreceptor-mediated vision in retinal degenerations: dark-adapted thresholds as outcome measures Exp Eye Res 80259s–272. [DOI] [PubMed] [Google Scholar]
- Ferris FL., 3rd, , Kassoff A, Bresnick GH., and, Bailey I. New visual acuity charts for clinical research. Am J Ophthalmol. 1982;94:91–96. [PubMed] [Google Scholar]
- Fishman GA, Jacobson SG, Alexander KR, Cideciyan AV, Birch DG, Weleber RG.et al. (2005Outcome measures and their application in clinical trials for retinal degenerative diseases: outline, review, and perspective Retina (Philadelphia, Pa) 25772–777. [DOI] [PubMed] [Google Scholar]
- Crossland MD, Culham LE, Kabanarou SA., and, Rubin GS. Preferred retinal locus development in patients with macular disease. Ophthalmology. 2005;112:1579–1585. doi: 10.1016/j.ophtha.2005.03.027. [DOI] [PubMed] [Google Scholar]
- Butz M, Wörgötter F., and, van Ooyen A. Activity-dependent structural plasticity. Brain Res Rev. 2009;60:287–305. doi: 10.1016/j.brainresrev.2008.12.023. [DOI] [PubMed] [Google Scholar]
- Déruaz A, Whatham AR, Mermoud C., and, Safran AB. Reading with multiple preferred retinal loci: implications for training a more efficient reading strategy. Vision Res. 2002;42:2947–2957. doi: 10.1016/s0042-6989(02)00354-1. [DOI] [PubMed] [Google Scholar]
- Bennett J, Ashtari M, Wellman J, Marshall KA, Cyckowski LL, Chung DC.et al. (2012AAV2 gene therapy readministration in three adults with congenital blindness Sci Transl Med 4120ra15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mackay DS, Henderson RH, Sergouniotis PI, Li Z, Moradi P, Holder GE.et al. (2010Novel mutations in MERTK associated with childhood onset rod-cone dystrophy Mol Vis 16369–377. [PMC free article] [PubMed] [Google Scholar]
- Shahzadi A, Riazuddin SA, Ali S, Li D, Khan SN, Husnain T.et al. (2010Nonsense mutation in MERTK causes autosomal recessive retinitis pigmentosa in a consanguineous Pakistani family Br J Ophthalmol 941094–1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ostergaard E, Duno M, Batbayli M, Vilhelmsen K., and, Rosenberg T. A novel MERTK deletion is a common founder mutation in the Faroe Islands and is responsible for a high proportion of retinitis pigmentosa cases. Mol Vis. 2011;17:1485–1492. [PMC free article] [PubMed] [Google Scholar]
- Bourne MC, Campbell DA., and, Pyke M. Cataract associated with an hereditary retinal lesion in rats. Br J Ophthalmol. 1938;22:608–613. doi: 10.1136/bjo.22.10.608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bourne MC, Campbell DA., and, Tansley K. Hereditary degeneration of the rat retina. Br J Ophthalmol. 1938;22:613–623. doi: 10.1136/bjo.22.10.613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dowling JE., and, Sidman RL. Inherited retinal dystrophy in the rat. J Cell Biol. 1962;14:73–109. doi: 10.1083/jcb.14.1.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herron WL, Riegel BW, Myers OE., and, Rubin ML. Retinal dystrophy in the rat–a pigment epithelial disease. Invest Ophthalmol. 1969;8:595–604. [PubMed] [Google Scholar]
- LaVail MM., and, Battelle BA. Influence of eye pigmentation and light deprivation on inherited retinal dystrophy in the rat. Exp Eye Res. 1975;21:167–192. doi: 10.1016/0014-4835(75)90080-9. [DOI] [PubMed] [Google Scholar]
- LaVail MM, Sidman M, Rausin R., and, Sidman RL. Discrimination of light intensity by rats with inherited retinal degeneration: a behavioral and cytological study. Vision Res. 1974;14:693–702. doi: 10.1016/0042-6989(74)90066-2. [DOI] [PubMed] [Google Scholar]
- D'Cruz PM, Yasumura D, Weir J, Matthes MT, Abderrahim H, LaVail MM.et al. (2000Mutation of the receptor tyrosine kinase gene Mertk in the retinal dystrophic RCS rat Hum Mol Genet 9645–651. [DOI] [PubMed] [Google Scholar]
- Gal A, Li Y, Thompson DA, Weir J, Orth U, Jacobson SG.et al. (2000Mutations in MERTK, the human orthologue of the RCS rat retinal dystrophy gene, cause retinitis pigmentosa Nat Genet 26270–271. [DOI] [PubMed] [Google Scholar]
- Vollrath D, Feng W, Duncan JL, Yasumura D, D'Cruz PM, Chappelow A.et al. (2001Correction of the retinal dystrophy phenotype of the RCS rat by viral gene transfer of Mertk Proc Natl Acad Sci USA 9812584–12589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith AJ, Schlichtenbrede FC, Tschernutter M, Bainbridge JW, Thrasher AJ., and, Ali RR. AAV-Mediated gene transfer slows photoreceptor loss in the RCS rat model of retinitis pigmentosa. Mol Ther. 2003;8:188–195. doi: 10.1016/s1525-0016(03)00144-8. [DOI] [PubMed] [Google Scholar]
- Tschernutter M, Schlichtenbrede FC, Howe S, Balaggan KS, Munro PM, Bainbridge JW.et al. (2005Long-term preservation of retinal function in the RCS rat model of retinitis pigmentosa following lentivirus-mediated gene therapy Gene Ther 12694–701. [DOI] [PubMed] [Google Scholar]
- Buch PK, MacLaren RE, Durán Y, Balaggan KS, MacNeil A, Schlichtenbrede FC.et al. (2006In contrast to AAV-mediated Cntf expression, AAV-mediated Gdnf expression enhances gene replacement therapy in rodent models of retinal degeneration Mol Ther 14700–709. [DOI] [PubMed] [Google Scholar]
- Deng WT, Dinculescu A, Li Q, Boye SL, Li J, Gorbatyuk MS.et al. (2012Tyrosine-mutant AAV8 delivery of human MERTK provides long-term retinal preservation in RCS rats Invest Ophthalmol Vis Sci 531895–1904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vernon M. Usher's syndrome–deafness and progressive blindness. Clinical cases, prevention, theory and literature survey. J Chronic Dis. 1969;22:133–151. doi: 10.1016/0021-9681(69)90055-1. [DOI] [PubMed] [Google Scholar]
- Boughman JA, Vernon M., and, Shaver KA. Usher syndrome: definition and estimate of prevalence from two high-risk populations. J Chronic Dis. 1983;36:595–603. doi: 10.1016/0021-9681(83)90147-9. [DOI] [PubMed] [Google Scholar]
- Petit C. Usher syndrome: from genetics to pathogenesis. Annu Rev Genomics Hum Genet. 2001;2:271–297. doi: 10.1146/annurev.genom.2.1.271. [DOI] [PubMed] [Google Scholar]
- Reiners J, Nagel-Wolfrum K, Jürgens K, Märker T., and, Wolfrum U. Molecular basis of human Usher syndrome: deciphering the meshes of the Usher protein network provides insights into the pathomechanisms of the Usher disease. Exp Eye Res. 2006;83:97–119. doi: 10.1016/j.exer.2005.11.010. [DOI] [PubMed] [Google Scholar]
- Sahly I, Dufour E, Schietroma C, Michel V, Bahloul A, Perfettini I.et al. (2012Localization of Usher 1 proteins to the photoreceptor calyceal processes, which are absent from mice J Cell Biol 199381–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weil D, Blanchard S, Kaplan J, Guilford P, Gibson F, Walsh J.et al. (1995Defective myosin VIIA gene responsible for Usher syndrome type 1B Nature 37460–61. [DOI] [PubMed] [Google Scholar]
- Smith RJ, Berlin CI, Hejtmancik JF, Keats BJ, Kimberling WJ, Lewis RA.et al. (1994Clinical diagnosis of the Usher syndromes. Usher Syndrome Consortium Am J Med Genet 5032–38. [DOI] [PubMed] [Google Scholar]
- Hasson T, Heintzelman MB, Santos-Sacchi J, Corey DP., and, Mooseker MS. Expression in cochlea and retina of myosin VIIa, the gene product defective in Usher syndrome type 1B. Proc Natl Acad Sci USA. 1995;92:9815–9819. doi: 10.1073/pnas.92.21.9815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weil D, Levy G, Sahly I, Levi-Acobas F, Blanchard S, El-Amraoui A.et al. (1996Human myosin VIIA responsible for the Usher 1B syndrome: a predicted membrane-associated motor protein expressed in developing sensory epithelia Proc Natl Acad Sci USA 933232–3237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu X, Vansant G, Udovichenko IP, Wolfrum U., and, Williams DS. Myosin VIIa, the product of the Usher 1B syndrome gene, is concentrated in the connecting cilia of photoreceptor cells. Cell Motil Cytoskeleton. 1997;37:240–252. doi: 10.1002/(SICI)1097-0169(1997)37:3<240::AID-CM6>3.0.CO;2-A. [DOI] [PubMed] [Google Scholar]
- Wolfrum U. The cellular function of the usher gene product myosin VIIa is specified by its ligands. Adv Exp Med Biol. 2003;533:133–142. doi: 10.1007/978-1-4615-0067-4_17. [DOI] [PubMed] [Google Scholar]
- Liu X, Ondek B., and, Williams DS. Mutant myosin VIIa causes defective melanosome distribution in the RPE of shaker-1 mice. Nat Genet. 1998;19:117–118. doi: 10.1038/470. [DOI] [PubMed] [Google Scholar]
- Gibbs D, Kitamoto J., and, Williams DS. Abnormal phagocytosis by retinal pigmented epithelium that lacks myosin VIIa, the Usher syndrome 1B protein. Proc Natl Acad Sci USA. 2003;100:6481–6486. doi: 10.1073/pnas.1130432100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gibbs D, Azarian SM, Lillo C, Kitamoto J, Klomp AE, Steel KP.et al. (2004Role of myosin VIIa and Rab27a in the motility and localization of RPE melanosomes J Cell Sci 117Pt 266473–6483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu X, Udovichenko IP, Brown SD, Steel KP., and, Williams DS. Myosin VIIa participates in opsin transport through the photoreceptor cilium. J Neurosci. 1999;19:6267–6274. doi: 10.1523/JNEUROSCI.19-15-06267.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gibbs D, Diemer T, Khanobdee K, Hu J, Bok D., and, Williams DS. Function of MYO7A in the human RPE and the validity of shaker1 mice as a model for Usher syndrome 1B. Invest Ophthalmol Vis Sci. 2010;51:1130–1135. doi: 10.1167/iovs.09-4032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hashimoto T, Gibbs D, Lillo C, Azarian SM, Legacki E, Zhang XM.et al. (2007Lentiviral gene replacement therapy of retinas in a mouse model for Usher syndrome type 1B Gene Ther 14584–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bainbridge JW, Stephens C, Parsley K, Demaison C, Halfyard A, Thrasher AJ.et al. (2001In vivo gene transfer to the mouse eye using an HIV-based lentiviral vector; efficient long-term transduction of corneal endothelium and retinal pigment epithelium Gene Ther 81665–1668. [DOI] [PubMed] [Google Scholar]
- Ikeda Y, Yonemitsu Y, Miyazaki M, Kohno R, Murakami Y, Murata T.et al. (2009Stable retinal gene expression in nonhuman primates via subretinal injection of SIVagm-based lentiviral vectors Hum Gene Ther 20573–579. [DOI] [PubMed] [Google Scholar]
- Jacobson SG, Cideciyan AV, Aleman TS, Sumaroka A, Roman AJ, Gardner LM.et al. (2008Usher syndromes due to MYO7A, PCDH15, USH2A or GPR98 mutations share retinal disease mechanism Hum Mol Genet 172405–2415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allocca M, Doria M, Petrillo M, Colella P, Garcia-Hoyos M, Gibbs D.et al. (2008Serotype-dependent packaging of large genes in adeno-associated viral vectors results in effective gene delivery in mice J Clin Invest 1181955–1964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu Z, Yang H., and, Colosi P. Effect of genome size on AAV vector packaging. Mol Ther. 2010;18:80–86. doi: 10.1038/mt.2009.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong B, Nakai H., and, Xiao W. Characterization of genome integrity for oversized recombinant AAV vector. Mol Ther. 2010;18:87–92. doi: 10.1038/mt.2009.258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lai Y, Yue Y., and, Duan D. Evidence for the failure of adeno-associated virus serotype 5 to package a viral genome > or = 8.2 kb. Mol Ther. 2010;18:75–79. doi: 10.1038/mt.2009.256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lopes VS, Boye SE, Boye, SL, Hauswirth WW, Williams DS.2010. XIV International Symposium on Retinal Degeneration. Vol. Mont-Tremblant, Quebec, Canada.
- Lopes VS, Diemer T, Gibbs, D, Boye SE, Boye SL, Hauswirth WW., and, Williams DS.2010. XIV International Symposium on Retinal Degeneration. Vol. Mont-Tremblant, Quebec, Canada.
- Allikmets R, Shroyer NF, Singh N, Seddon JM, Lewis RA, Bernstein PS.et al. (1997Mutation of the Stargardt disease gene (ABCR) in age-related macular degeneration Science 2771805–1807. [DOI] [PubMed] [Google Scholar]
- Quazi F, Lenevich S., and, Molday RS. ABCA4 is an N-retinylidene-phosphatidylethanolamine and phosphatidylethanolamine importer. Nat Commun. 2012;3:925. doi: 10.1038/ncomms1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Molday RS, Zhong M., and, Quazi F. The role of the photoreceptor ABC transporter ABCA4 in lipid transport and Stargardt macular degeneration. Biochim Biophys Acta. 2009;1791:573–583. doi: 10.1016/j.bbalip.2009.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mata NL, Weng J., and, Travis GH. Biosynthesis of a major lipofuscin fluorophore in mice and humans with ABCR-mediated retinal and macular degeneration. Proc Natl Acad Sci USA. 2000;97:7154–7159. doi: 10.1073/pnas.130110497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong J, Kim SR, Binley K, Pata I, Doi K, Mannik J.et al. (2008Correction of the disease phenotype in the mouse model of Stargardt disease by lentiviral gene therapy Gene Ther 151311–1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han Z, Conley SM, Makkia RS, Cooper MJ., and, Naash MI. DNA nanoparticle-mediated ABCA4 delivery rescues Stargardt dystrophy in mice. J Clin Invest. 2012;122:3221–3226. doi: 10.1172/JCI64833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MacDonald IM, Mah DY, Ho YK, Lewis RA., and, Seabra MC. A practical diagnostic test for choroideremia. Ophthalmology. 1998;105:1637–1640. doi: 10.1016/S0161-6420(98)99031-5. [DOI] [PubMed] [Google Scholar]
- Cremers FP, van de Pol DJ, van Kerkhoff LP, Wieringa B., and, Ropers HH. Cloning of a gene that is rearranged in patients with choroideraemia. Nature. 1990;347:674–677. doi: 10.1038/347674a0. [DOI] [PubMed] [Google Scholar]
- Seabra MC, Brown MS, Slaughter CA, Südhof TC., and, Goldstein JL. Purification of component A of Rab geranylgeranyl transferase: possible identity with the choroideremia gene product. Cell. 1992;70:1049–1057. doi: 10.1016/0092-8674(92)90253-9. [DOI] [PubMed] [Google Scholar]
- Seabra MC. New insights into the pathogenesis of choroideremia: a tale of two REPs. Ophthalmic Genet. 1996;17:43–46. doi: 10.3109/13816819609057869. [DOI] [PubMed] [Google Scholar]
- Kärnä J. Choroideremia. A clinical and genetic study of 84 Finnish patients and 126 female carriers. Acta Ophthalmol Suppl. 1986;176:1–68. [PubMed] [Google Scholar]
- Sorsby A, Franceschetti A, Joseph R, Davey JB. Choroideremia; clinical and genetic aspects. Br J Ophthalmol. 1952;36:547–581. doi: 10.1136/bjo.36.10.547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Cideciyan AV, Sumaroka A, Aleman TS, Schwartz SB, Windsor EA.et al. (2006Remodeling of the human retina in choroideremia: rab escort protein 1 (REP-1) mutations Invest Ophthalmol Vis Sci 474113–4120. [DOI] [PubMed] [Google Scholar]
- Zerial M., and, McBride H. Rab proteins as membrane organizers. Nat Rev Mol Cell Biol. 2001;2:107–117. doi: 10.1038/35052055. [DOI] [PubMed] [Google Scholar]
- Pfeffer SR. Rab GTPases: specifying and deciphering organelle identity and function. Trends Cell Biol. 2001;11:487–491. doi: 10.1016/s0962-8924(01)02147-x. [DOI] [PubMed] [Google Scholar]
- Pylypenko O, Rak A, Reents R, Niculae A, Sidorovitch V, Cioaca MD.et al. (2003Structure of Rab escort protein-1 in complex with Rab geranylgeranyltransferase Mol Cell 11483–494. [DOI] [PubMed] [Google Scholar]
- MacDonald IM, Sereda C, McTaggart K., and, Mah D. Choroideremia gene testing. Expert Rev Mol Diagn. 2004;4:478–484. doi: 10.1586/14737159.4.4.478. [DOI] [PubMed] [Google Scholar]
- Sergeev YV, Smaoui N, Sui R, Stiles D, Gordiyenko N, Strunnikova N.et al. (2009The functional effect of pathogenic mutations in Rab escort protein 1 Mutat Res 66544–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alory C., and, Balch WE. Organization of the Rab-GDI/CHM superfamily: the functional basis for choroideremia disease. Traffic. 2001;2:532–543. doi: 10.1034/j.1600-0854.2001.20803.x. [DOI] [PubMed] [Google Scholar]
- Gordiyenko NV, Fariss RN, Zhi C., and, MacDonald IM. Silencing of the CHM gene alters phagocytic and secretory pathways in the retinal pigment epithelium. Invest Ophthalmol Vis Sci. 2010;51:1143–1150. doi: 10.1167/iovs.09-4117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tolmachova T, Anders R, Abrink M, Bugeon L, Dallman MJ, Futter CE.et al. (2006Independent degeneration of photoreceptors and retinal pigment epithelium in conditional knockout mouse models of choroideremia J Clin Invest 116386–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tolmachova T, Wavre-Shapton ST, Barnard AR, MacLaren RE, Futter CE., and, Seabra MC. Retinal pigment epithelium defects accelerate photoreceptor degeneration in cell type-specific knockout mouse models of choroideremia. Invest Ophthalmol Vis Sci. 2010;51:4913–4920. doi: 10.1167/iovs.09-4892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anand V, Barral DC, Zeng Y, Brunsmann F, Maguire AM, Seabra MC.et al. (2003Gene therapy for choroideremia: in vitro rescue mediated by recombinant adenovirus Vision Res 43919–926. [DOI] [PubMed] [Google Scholar]
- Tolmachova T, Tolmachov OE, Wavre-Shapton ST, Tracey-White D, Futter CE., and, Seabra MC. CHM/REP1 cDNA delivery by lentiviral vectors provides functional expression of the transgene in the retinal pigment epithelium of choroideremia mice. J Gene Med. 2012;14:158–168. doi: 10.1002/jgm.1652. [DOI] [PubMed] [Google Scholar]
- MacLaren RE, Groppe M, Barnard AR, Tolmachova T, During MJ, Downes SM.et al (2012Association for Research in Vision and Opthalmology (ARVO), Ft. Lauderdale, FL
- Adamis AP, Miller JW, Bernal MT, D'Amico DJ, Folkman J, Yeo TK.et al. (1994Increased vascular endothelial growth factor levels in the vitreous of eyes with proliferative diabetic retinopathy Am J Ophthalmol 118445–450. [DOI] [PubMed] [Google Scholar]
- Kvanta A, Algvere PV, Berglin L., and, Seregard S. Subfoveal fibrovascular membranes in age-related macular degeneration express vascular endothelial growth factor. Invest Ophthalmol Vis Sci. 1996;37:1929–1934. [PubMed] [Google Scholar]
- Aiello LP, Pierce EA, Foley ED, Takagi H, Chen H, Riddle L.et al. (1995Suppression of retinal neovascularization in vivo by inhibition of vascular endothelial growth factor (VEGF) using soluble VEGF-receptor chimeric proteins Proc Natl Acad Sci USA 9210457–10461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown DM., and, Regillo CD. Anti-VEGF agents in the treatment of neovascular age-related macular degeneration: applying clinical trial results to the treatment of everyday patients. Am J Ophthalmol. 2007;144:627–637. doi: 10.1016/j.ajo.2007.06.039. [DOI] [PubMed] [Google Scholar]
- Rosenfeld PJ, Rich RM., and, Lalwani GA. Ranibizumab: Phase III clinical trial results. Ophthalmol Clin North Am. 2006;19:361–372. doi: 10.1016/j.ohc.2006.05.009. [DOI] [PubMed] [Google Scholar]
- D'Amico DJ, Masonson HN, Patel M, Adamis AP, Cunningham ET, Jr, Guyer DR.et al (2006Pegaptanib sodium for neovascular age-related macular degeneration: two-year safety results of the two prospective, multicenter, controlled clinical trials Ophthalmology 113992–1001. [DOI] [PubMed] [Google Scholar]
- Lai YK, Shen WY, Brankov M, Lai CM, Constable IJ., and, Rakoczy PE. Potential long-term inhibition of ocular neovascularisation by recombinant adeno-associated virus-mediated secretion gene therapy. Gene Ther. 2002;9:804–813. doi: 10.1038/sj.gt.3301695. [DOI] [PubMed] [Google Scholar]
- Lai CM, Shen WY, Brankov M, Lai YK, Barnett NL, Lee SY.et al. (2005Long-term evaluation of AAV-mediated sFlt-1 gene therapy for ocular neovascularization in mice and monkeys Mol Ther 12659–668. [DOI] [PubMed] [Google Scholar]
- Lai CM, Estcourt MJ, Himbeck RP, Lee SY, Yew-San Yeo I, Luu C.et al. (2012Preclinical safety evaluation of subretinal AAV2.sFlt-1 in non-human primates Gene Ther 19999–1009. [DOI] [PubMed] [Google Scholar]
- Pechan P, Rubin H, Lukason M, Ardinger J, DuFresne E, Hauswirth WW.et al. (2009Novel anti-VEGF chimeric molecules delivered by AAV vectors for inhibition of retinal neovascularization Gene Ther 1610–16. [DOI] [PubMed] [Google Scholar]
- Lai CM, Estcourt MJ, Wikstrom M, Himbeck RP, Barnett NL, Brankov M.et al. (2009rAAV.sFlt-1 gene therapy achieves lasting reversal of retinal neovascularization in the absence of a strong immune response to the viral vector Invest Ophthalmol Vis Sci 504279–4287. [DOI] [PubMed] [Google Scholar]
- Lukason M, DuFresne E, Rubin H, Pechan P, Li Q, Kim I.et al. (2011Inhibition of choroidal neovascularization in a nonhuman primate model by intravitreal administration of an AAV2 vector expressing a novel anti-VEGF molecule Mol Ther 19260–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Igarashi T, Miyake K, Masuda I, Takahashi H., and, Shimada T. Adeno-associated vector (type 8)-mediated expression of soluble Flt-1 efficiently inhibits neovascularization in a murine choroidal neovascularization model. Hum Gene Ther. 2010;21:631–637. doi: 10.1089/hum.2009.153. [DOI] [PubMed] [Google Scholar]
- Maclachlan TK, Lukason M, Collins M, Munger R, Isenberger E, Rogers C.et al. (2011Preclinical safety evaluation of AAV2-sFLT01- a gene therapy for age-related macular degeneration Mol Ther 19326–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O'Reilly MS, Boehm T, Shing Y, Fukai N, Vasios G, Lane WS.et al. (1997Endostatin: an endogenous inhibitor of angiogenesis and tumor growth Cell 88277–285. [DOI] [PubMed] [Google Scholar]
- O'Reilly MS. Angiostatin: an endogenous inhibitor of angiogenesis and of tumor growth. EXS. 1997;79:273–294. [PubMed] [Google Scholar]
- Mori K, Ando A, Gehlbach P, Nesbitt D, Takahashi K, Goldsteen D.et al. (2001Inhibition of choroidal neovascularization by intravenous injection of adenoviral vectors expressing secretable endostatin Am J Pathol 159313–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lai LJ, Xiao X., and, Wu JH. Inhibition of corneal neovascularization with endostatin delivered by adeno-associated viral (AAV) vector in a mouse corneal injury model. J Biomed Sci. 2007;14:313–322. doi: 10.1007/s11373-007-9153-7. [DOI] [PubMed] [Google Scholar]
- Raisler BJ, Berns KI, Grant MB, Beliaev D., and, Hauswirth WW. Adeno-associated virus type-2 expression of pigmented epithelium-derived factor or Kringles 1-3 of angiostatin reduce retinal neovascularization. Proc Natl Acad Sci USA. 2002;99:8909–8914. doi: 10.1073/pnas.122247299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Igarashi T, Miyake K, Kato K, Watanabe A, Ishizaki M, Ohara K.et al. (2003Lentivirus-mediated expression of angiostatin efficiently inhibits neovascularization in a murine proliferative retinopathy model Gene Ther 10219–226. [DOI] [PubMed] [Google Scholar]
- den Hollander AI, Roepman R, Koenekoop RK., and, Cremers FP. Leber congenital amaurosis: genes, proteins and disease mechanisms. Prog Retin Eye Res. 2008;27:391–419. doi: 10.1016/j.preteyeres.2008.05.003. [DOI] [PubMed] [Google Scholar]
- Milam AH, Barakat MR, Gupta N, Rose L, Aleman TS, Pianta MJ.et al. (2003Clinicopathologic effects of mutant GUCY2D in Leber congenital amaurosis Ophthalmology 110549–558. [DOI] [PubMed] [Google Scholar]
- Perrault I, Rozet JM, Gerber S, Ghazi I, Ducroq D, Souied E.et al. (2000Spectrum of retGC1 mutations in Leber's congenital amaurosis Eur J Hum Genet 8578–582. [DOI] [PubMed] [Google Scholar]
- Li L, Xiao X, Li S, Jia X, Wang P, Guo X.et al. (2011Detection of variants in 15 genes in 87 unrelated Chinese patients with Leber congenital amaurosis PLoS ONE 6e19458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dizhoor AM, Lowe DG, Olshevskaya EV, Laura RP., and, Hurley JB. The human photoreceptor membrane guanylyl cyclase, RetGC, is present in outer segments and is regulated by calcium and a soluble activator. Neuron. 1994;12:1345–1352. doi: 10.1016/0896-6273(94)90449-9. [DOI] [PubMed] [Google Scholar]
- Liu X, Seno K, Nishizawa Y, Hayashi F, Yamazaki A, Matsumoto H.et al. (1994Ultrastructural localization of retinal guanylate cyclase in human and monkey retinas Exp Eye Res 59761–768. [DOI] [PubMed] [Google Scholar]
- Polans A, Baehr W., and, Palczewski K. Turned on by Ca2+! The physiology and pathology of Ca(2+)-binding proteins in the retina. Trends Neurosci. 1996;19:547–554. doi: 10.1016/s0166-2236(96)10059-x. [DOI] [PubMed] [Google Scholar]
- Yang RB., and, Garbers DL. Two eye guanylyl cyclases are expressed in the same photoreceptor cells and form homomers in preference to heteromers. J Biol Chem. 1997;272:13738–13742. doi: 10.1074/jbc.272.21.13738. [DOI] [PubMed] [Google Scholar]
- Karan S, Frederick JM., and, Baehr W. Novel functions of photoreceptor guanylate cyclases revealed by targeted deletion. Mol Cell Biochem. 2010;334:141–155. doi: 10.1007/s11010-009-0322-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Cideciyan AV, Peshenko IV, Sumaroka A, Olshevskaya EV, Cao L.et al. (2013Determining consequences of retinal membrane guanylyl cyclase (RetGC1) deficiency in human Leber congenital amaurosis en route to therapy: residual cone-photoreceptor vision correlates with biochemical properties of the mutants Hum Mol Genet 22168–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pasadhika S, Fishman GA, Stone EM, Lindeman M, Zelkha R, Lopez I.et al. (2010Differential macular morphology in patients with RPE65-, CEP290-, GUCY2D-, and AIPL1-related Leber congenital amaurosis Invest Ophthalmol Vis Sci 512608–2614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simonelli F, Ziviello C, Testa F, Rossi S, Fazzi E, Bianchi PE.et al. (2007Clinical and molecular genetics of Leber's congenital amaurosis: a multicenter study of Italian patients Invest Ophthalmol Vis Sci 484284–4290. [DOI] [PubMed] [Google Scholar]
- Yang RB, Robinson SW, Xiong WH, Yau KW, Birch DG., and, Garbers DL. Disruption of a retinal guanylyl cyclase gene leads to cone-specific dystrophy and paradoxical rod behavior. J Neurosci. 1999;19:5889–5897. doi: 10.1523/JNEUROSCI.19-14-05889.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boye SE, Boye SL, Pang J, Ryals R, Everhart D, Umino Y.et al. (2010Functional and behavioral restoration of vision by gene therapy in the guanylate cyclase-1 (GC1) knockout mouse PLoS ONE 5e11306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boye SL, Conlon T, Erger K, Ryals R, Neeley A, Cossette T.et al. (2011Long-term preservation of cone photoreceptors and restoration of cone function by gene therapy in the guanylate cyclase-1 knockout (GC1KO) mouse Invest Ophthalmol Vis Sci 527098–7108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mihelec M, Pearson RA, Robbie SJ, Buch PK, Azam SA, Bainbridge JW.et al. (2011Long-term preservation of cones and improvement in visual function following gene therapy in a mouse model of leber congenital amaurosis caused by guanylate cyclase-1 deficiency Hum Gene Ther 221179–1190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baehr W, Karan S, Maeda T, Luo DG, Li S, Bronson JD.et al. (2007The function of guanylate cyclase 1 and guanylate cyclase 2 in rod and cone photoreceptors J Biol Chem 2828837–8847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boye SL, Peshenko IV, Huang WC, Min SH, McDoom I, Kay CN.et al. (2012. AAV-mediated gene therapy in the guanylate cyclase (RetGC1/RetGC2) double knockout mouse model of Leber congenital amaurosis. Hum Gene Ther (epub ahead of print). [DOI] [PMC free article] [PubMed]
- Boye SE, Alexander JJ, Boye SL, Witherspoon CD, Sandefer KJ, Conlon TJ.et al. (2012The human rhodopsin kinase promoter in an AAV5 vector confers rod- and cone-specific expression in the primate retina Hum Gene Ther 231101–1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thiadens AA, Somervuo V, van den Born LI, Roosing S, van Schooneveld MJ, Kuijpers RW.et al. (2010Progressive loss of cones in achromatopsia: an imaging study using spectral-domain optical coherence tomography Invest Ophthalmol Vis Sci 515952–5957. [DOI] [PubMed] [Google Scholar]
- Kohl S, Marx T, Giddings I, Jägle H, Jacobson SG, Apfelstedt-Sylla E.et al. (1998Total colourblindness is caused by mutations in the gene encoding the alpha-subunit of the cone photoreceptor cGMP-gated cation channel Nat Genet 19257–259. [DOI] [PubMed] [Google Scholar]
- Kohl S, Baumann B, Broghammer M, Jägle H, Sieving P, Kellner U.et al. (2000Mutations in the CNGB3 gene encoding the beta-subunit of the cone photoreceptor cGMP-gated channel are responsible for achromatopsia (ACHM3) linked to chromosome 8q21 Hum Mol Genet 92107–2116. [DOI] [PubMed] [Google Scholar]
- Aligianis IA, Forshew T, Johnson S, Michaelides M, Johnson CA, Trembath RC.et al. (2002Mapping of a novel locus for achromatopsia (ACHM4) to 1p and identification of a germline mutation in the alpha subunit of cone transducin (GNAT2) J Med Genet 39656–660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kohl S, Baumann B, Rosenberg T, Kellner U, Lorenz B, Vadalà M.et al. (2002Mutations in the cone photoreceptor G-protein alpha-subunit gene GNAT2 in patients with achromatopsia Am J Hum Genet 71422–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thiadens AA, den Hollander AI, Roosing S, Nabuurs SB, Zekveld-Vroon RC, Collin RW.et al. (2009Homozygosity mapping reveals PDE6C mutations in patients with early-onset cone photoreceptor disorders Am J Hum Genet 85240–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander JJ, Umino Y, Everhart D, Chang B, Min SH, Li Q.et al. (2007Restoration of cone vision in a mouse model of achromatopsia Nat Med 13685–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang B, Dacey MS, Hawes NL, Hitchcock PF, Milam AH, Atmaca-Sonmez P.et al. (2006Cone photoreceptor function loss-3, a novel mouse model of achromatopsia due to a mutation in Gnat2 Invest Ophthalmol Vis Sci 475017–5021. [DOI] [PubMed] [Google Scholar]
- Komáromy AM, Alexander JJ, Rowlan JS, Garcia MM, Chiodo VA, Kaya A.et al. (2010Gene therapy rescues cone function in congenital achromatopsia Hum Mol Genet 192581–2593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Michalakis S, Mühlfriedel R, Tanimoto N, Krishnamoorthy V, Koch S, Fischer MD.et al. (2010Restoration of cone vision in the CNGA3-/- mouse model of congenital complete lack of cone photoreceptor function Mol Ther 182057–2063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carvalho LS, Xu J, Pearson RA, Smith AJ, Bainbridge JW, Morris LM.et al. (2011Long-term and age-dependent restoration of visual function in a mouse model of CNGB3-associated achromatopsia following gene therapy Hum Mol Genet 203161–3175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pang JJ, Deng WT, Dai X, Lei B, Everhart D, Umino Y.et al. (2012AAV-mediated cone rescue in a naturally occurring mouse model of CNGA3-achromatopsia PLoS ONE 7e35250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- George ND, Yates JR., and, Moore AT. X linked retinoschisis. Br J Ophthalmol. 1995;79:697–702. doi: 10.1136/bjo.79.7.697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prenner JL, Capone A, Jr, Ciaccia S, Takada Y, Sieving PA., and, Trese MT. Congenital X-linked retinoschisis classification system. Retina (Philadelphia, Pa) 2006;26 suppl. 7:S61–S64. doi: 10.1097/01.iae.0000244290.09499.c1. [DOI] [PubMed] [Google Scholar]
- Minami Y, Ishiko S, Takai Y, Kato Y, Kagokawa H, Takamiya A.et al. (2005Retinal changes in juvenile X linked retinoschisis using three dimensional optical coherence tomography Br J Ophthalmol 891663–1664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peachey NS, Fishman GA, Derlacki DJ., and, Brigell MG. Psychophysical and electroretinographic findings in X-linked juvenile retinoschisis. Arch Ophthalmol. 1987;105:513–516. doi: 10.1001/archopht.1987.01060040083038. [DOI] [PubMed] [Google Scholar]
- Roesch MT, Ewing CC, Gibson AE., and, Weber BH. The natural history of X-linked retinoschisis. Can J Ophthalmol. 1998;33:149–158. [PubMed] [Google Scholar]
- Sauer CG, Gehrig A, Warneke-Wittstock R, Marquardt A, Ewing CC, Gibson A.et al. (1997Positional cloning of the gene associated with X-linked juvenile retinoschisis Nat Genet 17164–170. [DOI] [PubMed] [Google Scholar]
- Takada Y, Fariss RN, Muller M, Bush RA, Rushing EJ., and, Sieving PA. Retinoschisin expression and localization in rodent and human pineal and consequences of mouse RS1 gene knockout. Mol Vis. 2006;12:1108–1116. [PubMed] [Google Scholar]
- Wu WW, Wong JP, Kast J., and, Molday RS. RS1, a discoidin domain-containing retinal cell adhesion protein associated with X-linked retinoschisis, exists as a novel disulfide-linked octamer. J Biol Chem. 2005;280:10721–10730. doi: 10.1074/jbc.M413117200. [DOI] [PubMed] [Google Scholar]
- Baumgartner S, Hofmann K, Chiquet-Ehrismann R., and, Bucher P. The discoidin domain family revisited: new members from prokaryotes and a homology-based fold prediction. Protein Sci. 1998;7:1626–1631. doi: 10.1002/pro.5560070717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeng Y, Takada Y, Kjellstrom S, Hiriyanna K, Tanikawa A, Wawrousek E.et al. (2004RS-1 Gene Delivery to an Adult Rs1h Knockout Mouse Model Restores ERG b-Wave with Reversal of the Electronegative Waveform of X-Linked Retinoschisis Invest Ophthalmol Vis Sci 453279–3285. [DOI] [PubMed] [Google Scholar]
- Weber BH, Schrewe H, Molday LL, Gehrig A, White KL, Seeliger MW.et al. (2002Inactivation of the murine X-linked juvenile retinoschisis gene, Rs1h, suggests a role of retinoschisin in retinal cell layer organization and synaptic structure Proc Natl Acad Sci USA 996222–6227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kjellstrom S, Bush RA, Zeng Y, Takada Y., and, Sieving PA. Retinoschisin gene therapy and natural history in the Rs1h-KO mouse: long-term rescue from retinal degeneration. Invest Ophthalmol Vis Sci. 2007;48:3837–3845. doi: 10.1167/iovs.07-0203. [DOI] [PubMed] [Google Scholar]
- Min SH, Molday LL, Seeliger MW, Dinculescu A, Timmers AM, Janssen A.et al. (2005Prolonged recovery of retinal structure/function after gene therapy in an Rs1h-deficient mouse model of x-linked juvenile retinoschisis Mol Ther 12644–651. [DOI] [PubMed] [Google Scholar]
- Janssen A, Min SH, Molday LL, Tanimoto N, Seeliger MW, Hauswirth WW.et al. (2008Effect of late-stage therapy on disease progression in AAV-mediated rescue of photoreceptor cells in the retinoschisin-deficient mouse Mol Ther 161010–1017. [DOI] [PubMed] [Google Scholar]
- Takada Y, Vijayasarathy C, Zeng Y, Kjellstrom S, Bush RA., and, Sieving PA. Synaptic pathology in retinoschisis knockout (Rs1-/y) mouse retina and modification by rAAV-Rs1 gene delivery. Invest Ophthalmol Vis Sci. 2008;49:3677–3686. doi: 10.1167/iovs.07-1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park TK, Wu Z, Kjellstrom S, Zeng Y, Bush RA, Sieving PA.et al. (2009Intravitreal delivery of AAV8 retinoschisin results in cell type-specific gene expression and retinal rescue in the Rs1-KO mouse Gene Ther 16916–926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grayson C., and, Molday RS. Dominant negative mechanism underlies autosomal dominant Stargardt-like macular dystrophy linked to mutations in ELOVL4. J Biol Chem. 2005;280:32521–32530. doi: 10.1074/jbc.M503411200. [DOI] [PubMed] [Google Scholar]
- Sung CH, Makino C, Baylor D., and, Nathans J. A rhodopsin gene mutation responsible for autosomal dominant retinitis pigmentosa results in a protein that is defective in localization to the photoreceptor outer segment. J Neurosci. 1994;14:5818–5833. doi: 10.1523/JNEUROSCI.14-10-05818.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai X, Conley SM, Nash Z, Fliesler SJ, Cooper MJ., and, Naash MI. Gene delivery to mitotic and postmitotic photoreceptors via compacted DNA nanoparticles results in improved phenotype in a mouse model of retinitis pigmentosa. FASEB J. 2010;24:1178–1191. doi: 10.1096/fj.09-139147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LaVail MM, Yasumura D, Matthes MT, Drenser KA, Flannery JG, Lewin AS.et al. (2000Ribozyme rescue of photoreceptor cells in P23H transgenic rats: long-term survival and late-stage therapy Proc Natl Acad Sci USA 9711488–11493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lewin AS, Drenser KA, Hauswirth WW, Nishikawa S, Yasumura D, Flannery JG.et al. (1998Ribozyme rescue of photoreceptor cells in a transgenic rat model of autosomal dominant retinitis pigmentosa Nat Med 4967–971. [DOI] [PubMed] [Google Scholar]
- Millington-Ward S, O'Neill B, Tuohy G, Al-Jandal N, Kiang AS, Kenna PF.et al. (1997Strategems in vitro for gene therapies directed to dominant mutations Hum Mol Genet 61415–1426. [DOI] [PubMed] [Google Scholar]
- Chadderton N, Millington-Ward S, Palfi A, O'Reilly M, Tuohy G, Humphries MM.et al. (2009Improved retinal function in a mouse model of dominant retinitis pigmentosa following AAV-delivered gene therapy Mol Ther 17593–599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Georgiadis A, Tschernutter M, Bainbridge JW, Robbie SJ, McIntosh J, Nathwani AC.et al. (2010AAV-mediated knockdown of peripherin-2 in vivo using miRNA-based hairpins Gene Ther 17486–493. [DOI] [PubMed] [Google Scholar]
- Gorbatyuk M, Justilien V, Liu J, Hauswirth WW., and, Lewin AS. Preservation of photoreceptor morphology and function in P23H rats using an allele independent ribozyme. Exp Eye Res. 2007;84:44–52. doi: 10.1016/j.exer.2006.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorbatyuk MS, Pang JJ, Thomas J, Jr, Hauswirth WW., and, Lewin AS. Knockdown of wild-type mouse rhodopsin using an AAV vectored ribozyme as part of an RNA replacement approach. Mol Vis. 2005;11:648–656. [PubMed] [Google Scholar]
- Millington-Ward S, Chadderton N, O'Reilly M, Palfi A, Goldmann T, Kilty C.et al. (2011Suppression and replacement gene therapy for autosomal dominant disease in a murine model of dominant retinitis pigmentosa Mol Ther 19642–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O'Reilly M, Palfi A, Chadderton N, Millington-Ward S, Ader M, Cronin T.et al. (2007RNA interference-mediated suppression and replacement of human rhodopsin in vivo Am J Hum Genet 81127–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sullivan JM, Pietras KM, Shin BJ., and, Misasi JN. Hammerhead ribozymes designed to cleave all human rod opsin mRNAs which cause autosomal dominant retinitis pigmentosa. Mol Vis. 2002;8:102–113. [PubMed] [Google Scholar]
- Palfi A, Millington-Ward S, Chadderton N, O'Reilly M, Goldmann T, Humphries MM.et al. (2010Adeno-associated virus-mediated rhodopsin replacement provides therapeutic benefit in mice with a targeted disruption of the rhodopsin gene Hum Gene Ther 21311–323. [DOI] [PubMed] [Google Scholar]
- Mao H, Gorbatyuk MS, Rossmiller B, Hauswirth WW., and, Lewin AS. Long-term rescue of retinal structure and function by rhodopsin RNA replacement with a single adeno-associated viral vector in P23H RHO transgenic mice. Hum Gene Ther. 2012;23:356–366. doi: 10.1089/hum.2011.213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mussolino C, Sanges D, Marrocco E, Bonetti C, Di Vicino U, Marigo V.et al. (2011Zinc-finger-based transcriptional repression of rhodopsin in a model of dominant retinitis pigmentosa EMBO Mol Med 3118–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mao H, James T, Jr, Schwein A, Shabashvili AE, Hauswirth WW, Gorbatyuk MS.et al. (2011AAV delivery of wild-type rhodopsin preserves retinal function in a mouse model of autosomal dominant retinitis pigmentosa Hum Gene Ther 22567–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorbatyuk MS, Knox T, LaVail MM, Gorbatyuk OS, Noorwez SM, Hauswirth WW.et al. (2010Restoration of visual function in P23H rhodopsin transgenic rats by gene delivery of BiP/Grp78 Proc Natl Acad Sci USA 1075961–5966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wallace DC, Singh G, Lott MT, Hodge JA, Schurr TG, Lezza AM.et al. (1988Mitochondrial DNA mutation associated with Leber's hereditary optic neuropathy Science 2421427–1430. [DOI] [PubMed] [Google Scholar]
- Yu H, Koilkonda RD, Chou TH, Porciatti V, Ozdemir SS, Chiodo V.et al. (2012Gene delivery to mitochondria by targeting modified adenoassociated virus suppresses Leber's hereditary optic neuropathy in a mouse model Proc Natl Acad Sci USA 109E1238–E1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Glick B., and, Schatz G. Import of proteins into mitochondria. Annu Rev Genet. 1991;25:21–44. doi: 10.1146/annurev.ge.25.120191.000321. [DOI] [PubMed] [Google Scholar]
- Neupert W. Protein import into mitochondria. Annu Rev Biochem. 1997;66:863–917. doi: 10.1146/annurev.biochem.66.1.863. [DOI] [PubMed] [Google Scholar]
- Qi X, Sun L, Lewin AS, Hauswirth WW., and, Guy J. The mutant human ND4 subunit of complex I induces optic neuropathy in the mouse. Invest Ophthalmol Vis Sci. 2007;48:1–10. doi: 10.1167/iovs.06-0789. [DOI] [PubMed] [Google Scholar]
- Ellouze S, Augustin S, Bouaita A, Bonnet C, Simonutti M, Forster V.et al. (2008Optimized allotopic expression of the human mitochondrial ND4 prevents blindness in a rat model of mitochondrial dysfunction Am J Hum Genet 83373–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guy J, Qi X, Koilkonda RD, Arguello T, Chou TH, Ruggeri M.et al. (2009Efficiency and safety of AAV-mediated gene delivery of the human ND4 complex I subunit in the mouse visual system Invest Ophthalmol Vis Sci 504205–4214. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.