Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Pathol ; 193(11): 1694-1705, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37330004

ABSTRACT

CreTrp1 mice are widely used for conditional retinal pigment epithelium (RPE) gene function studies. Like other Cre/LoxP models, phenotypes in CreTrp1 mice can be affected by Cre-mediated cellular toxicity, leading to RPE dysfunction, altered morphology and atrophy, activation of innate immunity, and consequent impairment of photoreceptor function. These effects are common among the age-related alterations of RPE that feature in early/intermediate forms of age-related macular degeneration. This article characterizes Cre-mediated pathology in the CreTrp1 line to elucidate the impact of RPE degeneration on both developmental and pathologic choroidal neovascularization. Nonredundant roles of the two major components of the hypoxia-inducible factor (HIF) family of transcription regulators, HIF1α and HIF2α, were identified. Genetic ablation of Hif1a protected against Cre-induced degeneration of RPE and choroid, whereas ablation of Hif2a exacerbated this degeneration. Furthermore, HIF1α deficiency protected CreTrp1 mice against laser-induced choroidal neovascularization, whereas HIF2α deficiency exacerbated the phenotype. Cre-mediated degeneration of the RPE in CreTrp1 mice offers an opportunity to investigate the impact of hypoxia signaling in the context of RPE degeneration. These findings indicate that HIF1α promotes Cre recombinase-mediated RPE degeneration and laser-induced choroidal neovascularization, whereas HIF2α is protective.

2.
J Biol Chem ; 298(6): 101944, 2022 06.
Article in English | MEDLINE | ID: mdl-35447116

ABSTRACT

Mechanistic target of rapamycin (mTOR) and mTOR complex 1 (mTORC1), linchpins of the nutrient sensing and protein synthesis pathways, are present at relatively high levels in the ganglion cell layer (GCL) and retinal ganglion cells (RGCs) of rodent and human retinas. However, the role of mTORCs in the control of protein synthesis in RGC is unknown. Here, we applied the SUrface SEnsing of Translation (SUnSET) method of nascent protein labeling to localize and quantify protein synthesis in the retinas of adult mice. We also used intravitreal injection of an adeno-associated virus 2 vector encoding Cre recombinase in the eyes of mtor- or rptor-floxed mice to conditionally knockout either both mTORCs or only mTORC1, respectively, in cells within the GCL. A novel vector encoding an inactive Cre mutant (CreΔC) served as control. We found that retinal protein synthesis was highest in the GCL, particularly in RGC. Negation of both complexes or only mTORC1 significantly reduced protein synthesis in RGC. In addition, loss of mTORC1 function caused a significant reduction in the pan-RGC marker, RNA-binding protein with multiple splicing, with little decrease of the total number of cells in the RGC layer, even at 25 weeks after adeno-associated virus-Cre injection. These findings reveal that mTORC1 signaling is necessary for maintaining the high rate of protein synthesis in RGCs of adult rodents, but it may not be essential to maintain RGC viability. These findings may also be relevant to understanding the pathophysiology of RGC disorders, including glaucoma, diabetic retinopathy, and optic neuropathies.


Subject(s)
Glaucoma , Retinal Ganglion Cells , Animals , Glaucoma/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Retina/metabolism , Retinal Ganglion Cells/metabolism
3.
Hum Mol Genet ; 29(22): 3706-3716, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33355362

ABSTRACT

Mutations in retinitis pigmentosa GTPase regulator (RPGR) cause severe retinal ciliopathy, X-linked retinitis pigmentosa. Although two major alternatively spliced isoforms, RPGRex1-19 and RPGRORF15, are expressed, the relative importance of these isoforms in disease pathogenesis is unclear. Here, we analyzed fibroblast samples from eight patients and found that all of them form longer cilia than normal controls, albeit to different degrees. Although all mutant RPGRORF15 messenger RNAs (mRNAs) are unstable, their steady-state levels were similar or higher than those in the control cells, suggesting there may be increased transcription. Three of the fibroblasts that had higher levels of mutant RPGRORF15 mRNA also exhibited significantly higher levels of RPGRex1-19 mRNA. Four samples with unaltered RPGRex1-19 levels carried mutations in RPGRORF15 that resulted in this isoform being relatively less stable. Thus, in all cases, the RPGRex1-19/RPGRORF15 isoform ratio was increased, and this was highly correlative to the cilia extension defect. Moreover, overexpression of RPGRex1-19 (mimicking the increase in RPGRex1-19 to RPGRORF15 isoform ratio) or RPGRORF15 (mimicking reduction of the ratio) resulted in significantly longer or shorter cilia, respectively. Notably, the cilia length defect appears to be attributable to both the loss of the wild-type RPGRORF15 protein and to the higher levels of the RPGRex1-19 isoform, indicating that the observed defect is due to the altered isoform ratios. These results suggest that maintaining the optimal RPGRex1-9 to RPGRORF15 ratio is critical for cilia growth and that designing strategies that focus on the best ways to restore the RPGRex1-19/RPGRORF15 ratio may lead to better therapeutic outcomes.


Subject(s)
Eye Proteins/genetics , Genetic Diseases, X-Linked/genetics , Retinitis Pigmentosa/genetics , Alternative Splicing/genetics , Carrier Proteins/genetics , Cilia/genetics , Cilia/pathology , Exons/genetics , Female , Fibroblasts , Genetic Diseases, X-Linked/pathology , Humans , Male , Mutation/genetics , Protein Isoforms/genetics , Retina/metabolism , Retina/pathology , Retinitis Pigmentosa/pathology
4.
Development ; 147(18)2020 09 28.
Article in English | MEDLINE | ID: mdl-32878923

ABSTRACT

Multipotent retinal progenitor cells (RPCs) generate various cell types in a precise chronological order, but how exactly cone photoreceptor production is restricted to early stages remains unclear. Here, we show that the POU-homeodomain factors Pou2f1/Pou2f2, the homologs of Drosophila temporal identity factors nub/pdm2, regulate the timely production of cones in mice. Forcing sustained expression of Pou2f1 or Pou2f2 in RPCs expands the period of cone production, whereas misexpression in late-stage RPCs triggers ectopic cone production at the expense of late-born fates. Mechanistically, we report that Pou2f1 induces Pou2f2 expression, which binds to a POU motif in the promoter of the rod-inducing factor Nrl to repress its expression. Conversely, conditional inactivation of Pou2f2 in RPCs increases Nrl expression and reduces cone production. Finally, we provide evidence that Pou2f1 is part of a cross-regulatory cascade with the other temporal identity factors Ikzf1 and Casz1. These results uncover Pou2f1/2 as regulators of the temporal window for cone genesis and, given their widespread expression in the nervous system, raise the possibility of a general role in temporal patterning.This article has an associated 'The people behind the papers' interview.


Subject(s)
Eye Proteins/metabolism , Octamer Transcription Factor-1/metabolism , Octamer Transcription Factor-2/metabolism , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Animals , Drosophila/metabolism , Female , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Retinal Rod Photoreceptor Cells/metabolism , Stem Cells/metabolism
5.
EMBO Rep ; 22(11): e53732, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34494703

ABSTRACT

Neuronal communication is typically mediated via synapses and gap junctions. New forms of intercellular communication, including nanotubes (NTs) and extracellular vesicles (EVs), have been described for non-neuronal cells, but their role in neuronal communication is not known. Recently, transfer of cytoplasmic material between donor and host neurons ("material transfer") was shown to occur after photoreceptor transplantation. The cellular mechanism(s) underlying this surprising finding are unknown. Here, using transplantation, primary neuronal cultures and the generation of chimeric retinae, we show for the first time that mammalian photoreceptor neurons can form open-end NT-like processes. These processes permit the transfer of cytoplasmic and membrane-bound molecules in culture and after transplantation and can mediate gain-of-function in the acceptor cells. Rarely, organelles were also observed to transfer. Strikingly, use of chimeric retinae revealed that material transfer can occur between photoreceptors in the intact adult retina. Conversely, while photoreceptors are capable of releasing EVs, at least in culture, these are taken up by glia and not by retinal neurons. Our findings provide the first evidence of functional NT-like processes forming between sensory neurons in culture and in vivo.


Subject(s)
Extracellular Vesicles , Nanotubes , Animals , Cell Communication , Mammals , Neurons , Retina
6.
Hum Mol Genet ; 29(8): 1310-1318, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32196553

ABSTRACT

Rhodopsin misfolding caused by the P23H mutation is a major cause of autosomal dominant retinitis pigmentosa (adRP). To date, there are no effective treatments for adRP. The BiP co-chaperone and reductase ERdj5 (DNAJC10) is part of the endoplasmic reticulum (ER) quality control machinery, and previous studies have shown that overexpression of ERdj5 in vitro enhanced the degradation of P23H rhodopsin, whereas knockdown of ERdj5 increased P23H rhodopsin ER retention and aggregation. Here, we investigated the role of ERdj5 in photoreceptor homeostasis in vivo by using an Erdj5 knockout mouse crossed with the P23H knock-in mouse and by adeno-associated viral (AAV) vector-mediated gene augmentation of ERdj5 in P23H-3 rats. Electroretinogram (ERG) and optical coherence tomography of Erdj5-/- and P23H+/-:Erdj5-/- mice showed no effect of ERdj5 ablation on retinal function or photoreceptor survival. Rhodopsin levels and localization were similar to those of control animals at a range of time points. By contrast, when AAV2/8-ERdj5-HA was subretinally injected into P23H-3 rats, analysis of the full-field ERG suggested that overexpression of ERdj5 reduced visual function loss 10 weeks post-injection (PI). This correlated with a significant preservation of photoreceptor cells at 4 and 10 weeks PI. Assessment of the outer nuclear layer (ONL) morphology showed preserved ONL thickness and reduced rhodopsin retention in the ONL in the injected superior retina. Overall, these data suggest that manipulation of the ER quality control and ER-associated degradation factors to promote mutant protein degradation could be beneficial for the treatment of adRP caused by mutant rhodopsin.


Subject(s)
HSP40 Heat-Shock Proteins/genetics , Molecular Chaperones/genetics , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , Animals , Disease Models, Animal , Electroretinography , Endoplasmic Reticulum/genetics , Gene Knock-In Techniques , Mice , Mice, Knockout , Mutation/genetics , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Rats , Retina/metabolism , Retina/pathology , Retinitis Pigmentosa/pathology , Rhodopsin/metabolism , Transfection
7.
Exp Eye Res ; 215: 108908, 2022 02.
Article in English | MEDLINE | ID: mdl-34954204

ABSTRACT

Opticin is an extracellular glycoprotein present in the vitreous. Its antiangiogenic properties offer the potential for therapeutic intervention in conditions such as proliferative diabetic retinopathy and retinopathy of prematurity. Here, we investigated the hypothesis that intravitreal administration of recombinant human opticin can safely protect against the development of pathological angiogenesis and promote its regression. We generated and purified recombinant human opticin and investigated its impact on the development and regression of pathological retinal neovascularization following intravitreal administration in murine oxygen-induced retinopathy. We also investigated its effect on normal retinal vascular development and function, following intravitreal injection in neonatal mice, by histological examination and electroretinography. In oxygen-induced retinopathy, intravitreal administration of human recombinant opticin protected against the development of retinal neovascularization to similar extent as aflibercept, which targets VEGF. Opticin also accelerated regression of established retinal neovascularization, though the effect at 18 h was less than that of aflibercept. Intravitreal administration of human recombinant opticin in neonatal mice caused no detectable perturbation of subsequent retinal vascular development or function. In summary we found that intraocular administration of recombinant human opticin protects against the development of pathological angiogenesis in mice and promotes its regression.


Subject(s)
Hyperoxia , Retinal Neovascularization , Retinopathy of Prematurity , Animals , Disease Models, Animal , Humans , Hyperoxia/complications , Infant, Newborn , Intravitreal Injections , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , Oxygen/toxicity , Retinal Neovascularization/drug therapy , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/prevention & control
8.
Glia ; 69(9): 2272-2290, 2021 09.
Article in English | MEDLINE | ID: mdl-34029407

ABSTRACT

Gliosis is a complex process comprising upregulation of intermediate filament (IF) proteins, particularly glial fibrillary acidic protein (GFAP) and vimentin, changes in glial cell morphology (hypertrophy) and increased deposition of inhibitory extracellular matrix molecules. Gliosis is common to numerous pathologies and can have deleterious effects on tissue function and regeneration. The role of IFs in gliosis is controversial, but a key hypothesized function is the stabilization of glial cell hypertrophy. Here, we developed RNAi approaches to examine the role of GFAP and vimentin in vivo in a murine model of inherited retinal degeneration, the Rhodopsin knockout (Rho-/- ) mouse. Specifically, we sought to examine the role of these IFs in the establishment of Müller glial hypertrophy during progressive degeneration, as opposed to (more commonly assessed) acute injury. Prevention of Gfap upregulation had a significant effect on the morphology of reactive Müller glia cells in vivo and, more strikingly, the reduction of Vimentin expression almost completely prevented these cells from undergoing degeneration-associated hypertrophy. Moreover, and in contrast to studies in knockout mice, simultaneous suppression of both GFAP and vimentin expression led to severe changes in the cytoarchitecture of the retina, in both diseased and wild-type eyes. These data demonstrate a crucial role for Vimentin, as well as GFAP, in the establishment of glial hypertrophy and support the further exploration of RNAi-mediated knockdown of vimentin as a potential therapeutic approach for modulating scar formation in the degenerating retina.


Subject(s)
Ependymoglial Cells , Glial Fibrillary Acidic Protein , Retinal Degeneration , Vimentin , Animals , Ependymoglial Cells/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hypertrophy/metabolism , Hypertrophy/pathology , Intermediate Filaments/metabolism , Mice , Neuroglia/metabolism , RNA Interference , Retina/metabolism , Retinal Degeneration/pathology , Vimentin/metabolism
9.
Hum Mol Genet ; 28(23): 3867-3879, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31807779

ABSTRACT

The neuronal ceroid lipofuscinoses (NCLs), more commonly referred to as Batten disease, are a group of inherited lysosomal storage disorders that present with neurodegeneration, loss of vision and premature death. There are at least 13 genetically distinct forms of NCL. Enzyme replacement therapies and pre-clinical studies on gene supplementation have shown promising results for NCLs caused by lysosomal enzyme deficiencies. The development of gene therapies targeting the brain for NCLs caused by defects in transmembrane proteins has been more challenging and only limited therapeutic effects in animal models have been achieved so far. Here, we describe the development of an adeno-associated virus (AAV)-mediated gene therapy to treat the neurodegeneration in a mouse model of CLN6 disease, a form of NCL with a deficiency in the membrane-bound protein CLN6. We show that neonatal bilateral intracerebroventricular injections with AAV9 carrying CLN6 increase lifespan by more than 90%, maintain motor skills and motor coordination and reduce neuropathological hallmarks of Cln6-deficient mice up to 23 months post vector administration. These data demonstrate that brain-directed gene therapy is a valid strategy to treat the neurodegeneration of CLN6 disease and may be applied to other forms of NCL caused by transmembrane protein deficiencies in the future.


Subject(s)
Genetic Vectors/administration & dosage , Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/therapy , Animals , Animals, Newborn , Brain/growth & development , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy , Humans , Injections, Intraventricular , Membrane Proteins/metabolism , Mice , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/metabolism , Treatment Outcome
10.
Development ; 145(8)2018 04 25.
Article in English | MEDLINE | ID: mdl-29615467

ABSTRACT

In the adult central nervous system, endothelial and neuronal cells engage in tight cross-talk as key components of the so-called neurovascular unit. Impairment of this important relationship adversely affects tissue homeostasis, as observed in neurodegenerative conditions including Alzheimer's and Parkinson's disease. In development, the influence of neuroprogenitor cells on angiogenesis is poorly understood. Here, we show in mouse that these cells interact intimately with the growing retinal vascular network, and we identify a novel regulatory mechanism of vasculature development mediated by hypoxia-inducible factor 2a (Hif2a). By Cre-lox gene excision, we show that Hif2a in retinal neuroprogenitor cells upregulates the expression of the pro-angiogenic mediators vascular endothelial growth factor and erythropoietin, whereas it locally downregulates the angiogenesis inhibitor endostatin. Importantly, absence of Hif2a in retinal neuroprogenitor cells causes a marked reduction of proliferating endothelial cells at the angiogenic front. This results in delayed retinal vascular development, fewer major retinal vessels and reduced density of the peripheral deep retinal vascular plexus. Our findings demonstrate that retinal neuroprogenitor cells are a crucial component of the developing neurovascular unit.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Retinal Vessels/growth & development , Retinal Vessels/innervation , Animals , Astrocytes/cytology , Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation , Endostatins/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neovascularization, Physiologic/genetics , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Retinal Pigment Epithelium/growth & development , Retinal Pigment Epithelium/metabolism , Retinal Vessels/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
Mol Ther ; 28(3): 820-829, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31981492

ABSTRACT

Glaucoma is a common cause of blindness, yet current therapeutic options are imperfect. Clinical trials have invariably shown that reduction in intraocular pressure (IOP) regardless of disease subtype prevents visual loss. Reducing ciliary body aqueous humor production can lower IOP, and the adeno-associated virus ShH10 serotype was identified as able to transduce mouse ciliary body epithelium following intravitreal injection. Using ShH10 to deliver a single vector CRISPR-Cas9 system disrupting Aquaporin 1 resulted in reduced IOP in treated eyes (10.4 ± 2.4 mmHg) compared with control (13.2 ± 2.0 mmHg) or non-injected eyes (13.1 ± 2.8 mmHg; p < 0.001; n = 12). Editing in the aquaporin 1 gene could be detected in ciliary body, and no off-target increases in corneal or retinal thickness were identified. In experimental mouse models of corticosteroid and microbead-induced ocular hypertension, IOP could be reduced to prevent ganglion cell loss (32 ± 4 /mm2) compared with untreated eyes (25 ± 5/mm2; p < 0.01). ShH10 could transduce human ciliary body from post-mortem donor eyes in ex vivo culture with indel formation detectable in the Aquaporin 1 locus. Clinical translation of this approach to patients with glaucoma may permit long-term reduction of IOP following a single injection.


Subject(s)
Aquaporin 1/genetics , Ciliary Body/metabolism , Gene Editing , Genetic Therapy , Glaucoma/genetics , Glaucoma/therapy , Animals , Aquaporin 1/metabolism , Base Sequence , CRISPR-Cas Systems , Dependovirus/genetics , Gene Expression , Gene Targeting , Genetic Therapy/methods , Genetic Vectors/genetics , Glaucoma/diagnosis , Glaucoma/physiopathology , Mice , Retina/metabolism , Retina/pathology , Transduction, Genetic , Transgenes
12.
Angiogenesis ; 23(2): 83-90, 2020 05.
Article in English | MEDLINE | ID: mdl-31583505

ABSTRACT

The retinal vasculature is tightly organized in a structure that provides for the high metabolic demand of neurons while minimizing interference with incident light. The adverse impact of retinal vascular insufficiency is mitigated by adaptive vascular regeneration but exacerbated by pathological neovascularization. Aberrant growth of neovessels in the retina is responsible for impairment of sight in common blinding disorders including retinopathy of prematurity, proliferative diabetic retinopathy, and age-related macular degeneration. Myeloid cells are key players in this process, with diverse roles that can either promote or protect against ocular neovascularization. We have previously demonstrated that myeloid-derived VEGF, HIF1, and HIF2 are not essential for pathological retinal neovascularization. Here, however, we show by cell-specific depletion of Vhl in a mouse model of retinal ischemia (oxygen-induced retinopathy, OIR) that myeloid-derived HIFs promote VEGF and bFGF expression and enhance vascular regeneration in association with improved density and organization of the astrocytic network.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Ischemia/genetics , Myeloid Cells/metabolism , Regeneration/genetics , Retinal Vessels/physiology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia/genetics , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Transgenic , Retina/pathology , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Diseases/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
13.
Hum Mol Genet ; 26(2): 305-319, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28065882

ABSTRACT

Protein misfolding caused by inherited mutations leads to loss of protein function and potentially toxic 'gain of function', such as the dominant P23H rhodopsin mutation that causes retinitis pigmentosa (RP). Here, we tested whether the AMPK activator metformin could affect the P23H rhodopsin synthesis and folding. In cell models, metformin treatment improved P23H rhodopsin folding and traffic. In animal models of P23H RP, metformin treatment successfully enhanced P23H traffic to the rod outer segment, but this led to reduced photoreceptor function and increased photoreceptor cell death. The metformin-rescued P23H rhodopsin was still intrinsically unstable and led to increased structural instability of the rod outer segments. These data suggest that improving the traffic of misfolding rhodopsin mutants is unlikely to be a practical therapy, because of their intrinsic instability and long half-life in the outer segment, but also highlights the potential of altering translation through AMPK to improve protein function in other protein misfolding diseases.


Subject(s)
AMP-Activated Protein Kinases/genetics , Metformin/administration & dosage , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , AMP-Activated Protein Kinases/biosynthesis , Animals , Disease Models, Animal , Humans , Mice , Mutant Proteins/genetics , Photoreceptor Cells/drug effects , Photoreceptor Cells/pathology , Protein Folding/drug effects , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/pathology , Rats , Retinal Degeneration/drug therapy , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/pathology , Rhodopsin/chemistry , Rod Cell Outer Segment/drug effects , Rod Cell Outer Segment/pathology , Transcriptional Activation/drug effects
14.
Stem Cells ; 36(5): 709-722, 2018 05.
Article in English | MEDLINE | ID: mdl-29327488

ABSTRACT

Loss of photoreceptor cells due to retinal degeneration is one of the main causes of blindness in the developed world. Although there is currently no effective treatment, cell replacement therapy using stem-cell-derived photoreceptor cells may be a feasible future treatment option. In order to ensure safety and efficacy of this approach, robust cell isolation and purification protocols must be developed. To this end, we previously developed a biomarker panel for the isolation of mouse photoreceptor precursors from the developing mouse retina and mouse embryonic stem cell cultures. In the current study we applied this approach to the human pluripotent stem cell (hPSC) system, and identified novel biomarker combinations that can be leveraged for the isolation of human photoreceptors. Human retinal samples and hPSC-derived retinal organoid cultures were screened against 242 human monoclonal antibodies using a high through-put flow cytometry approach. We identified 46 biomarkers with significant expression levels in the human retina and hPSC differentiation cultures. Human retinal cell samples, either from fetal tissue or derived from embryonic and induced pluripotent stem cell cultures, were fluorescence-activated cell sorted (FACS) using selected candidate biomarkers that showed expression in discrete cell populations. Enrichment for photoreceptors and exclusion of mitotically active cells was demonstrated by immunocytochemical analysis with photoreceptor-specific antibodies and Ki-67. We established a biomarker combination, which enables the robust purification of viable human photoreceptors from both human retinae and hPSC-derived organoid cultures. Stem Cells 2018;36:709-722.


Subject(s)
Cell Differentiation/physiology , Induced Pluripotent Stem Cells/cytology , Photoreceptor Cells/cytology , Retinal Degeneration/therapy , Animals , Biomarkers/analysis , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Photoreceptor Cells, Vertebrate/cytology , Pluripotent Stem Cells/cytology , Stem Cell Transplantation/methods
15.
Brain ; 141(7): 2014-2031, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29788236

ABSTRACT

Recombinant adeno-associated viruses (AAVs) are popular in vivo gene transfer vehicles. However, vector doses needed to achieve therapeutic effect are high and some target tissues in the central nervous system remain difficult to transduce. Gene therapy trials using AAV for the treatment of neurological disorders have seldom led to demonstrated clinical efficacy. Important contributing factors are low transduction rates and inefficient distribution of the vector. To overcome these hurdles, a variety of capsid engineering methods have been utilized to generate capsids with improved transduction properties. Here we describe an alternative approach to capsid engineering, which draws on the natural evolution of the virus and aims to yield capsids that are better suited to infect human tissues. We generated an AAV capsid to include amino acids that are conserved among natural AAV2 isolates and tested its biodistribution properties in mice and rats. Intriguingly, this novel variant, AAV-TT, demonstrates strong neurotropism in rodents and displays significantly improved distribution throughout the central nervous system as compared to AAV2. Additionally, sub-retinal injections in mice revealed markedly enhanced transduction of photoreceptor cells when compared to AAV2. Importantly, AAV-TT exceeds the distribution abilities of benchmark neurotropic serotypes AAV9 and AAVrh10 in the central nervous system of mice, and is the only virus, when administered at low dose, that is able to correct the neurological phenotype in a mouse model of mucopolysaccharidosis IIIC, a transmembrane enzyme lysosomal storage disease, which requires delivery to every cell for biochemical correction. These data represent unprecedented correction of a lysosomal transmembrane enzyme deficiency in mice and suggest that AAV-TT-based gene therapies may be suitable for treatment of human neurological diseases such as mucopolysaccharidosis IIIC, which is characterized by global neuropathology.


Subject(s)
Capsid/physiology , Genetic Therapy/methods , Protein Engineering/methods , Animals , Dependovirus/genetics , Female , Genetic Vectors , Male , Mice , Mice, Inbred C57BL , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/therapy , Photoreceptor Cells/drug effects , Rats , Rats, Sprague-Dawley , Retina/physiology , Tissue Distribution , Transduction, Genetic
16.
Mol Ther ; 26(5): 1343-1353, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29606505

ABSTRACT

The neuronal ceroid lipofuscinoses (NCLs) are inherited lysosomal storage disorders characterized by general neurodegeneration and premature death. Sight loss is also a major symptom in NCLs, severely affecting the quality of life of patients, but it is not targeted effectively by brain-directed therapies. Here we set out to explore the therapeutic potential of an ocular gene therapy to treat sight loss in NCL due to a deficiency in the transmembrane protein CLN6. We found that, although Cln6nclf mice presented mainly with photoreceptor degeneration, supplementation of CLN6 in photoreceptors was not beneficial. Because the level of CLN6 is low in photoreceptors but high in bipolar cells (retinal interneurons that are only lost in Cln6-deficient mice at late disease stages), we explored the therapeutic effects of delivering CLN6 to bipolar cells using adeno-associated virus (AAV) serotype 7m8. Bipolar cell-specific expression of CLN6 slowed significantly the loss of photoreceptor function and photoreceptor cells. This study shows that the deficiency of a gene normally expressed in bipolar cells can cause the loss of photoreceptors and that this can be prevented by bipolar cell-directed treatment.


Subject(s)
Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Photoreceptor Cells/metabolism , Retinal Bipolar Cells/metabolism , Animals , Dependovirus/genetics , Disease Models, Animal , Gene Expression , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/genetics , Humans , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Neuronal Ceroid-Lipofuscinoses/metabolism , Neuronal Ceroid-Lipofuscinoses/pathology , Neuronal Ceroid-Lipofuscinoses/therapy , Photoreceptor Cells/pathology
17.
N Engl J Med ; 372(20): 1887-97, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25938638

ABSTRACT

BACKGROUND: Mutations in RPE65 cause Leber's congenital amaurosis, a progressive retinal degenerative disease that severely impairs sight in children. Gene therapy can result in modest improvements in night vision, but knowledge of its efficacy in humans is limited. METHODS: We performed a phase 1-2 open-label trial involving 12 participants to evaluate the safety and efficacy of gene therapy with a recombinant adeno-associated virus 2/2 (rAAV2/2) vector carrying the RPE65 complementary DNA, and measured visual function over the course of 3 years. Four participants were administered a lower dose of the vector, and 8 were administered a higher dose. In a parallel study in dogs, we investigated the relationship among vector dose, visual function, and electroretinography (ERG) findings. RESULTS: Improvements in retinal sensitivity were evident, to varying extents, in six participants for up to 3 years, peaking at 6 to 12 months after treatment and then declining. No associated improvement in retinal function was detected by means of ERG. Three participants had intraocular inflammation, and two had clinically significant deterioration of visual acuity. The reduction in central retinal thickness varied among participants. In dogs, RPE65 gene therapy with the same vector at lower doses improved vision-guided behavior, but only higher doses resulted in improvements in retinal function that were detectable with the use of ERG. CONCLUSIONS: Gene therapy with rAAV2/2 RPE65 vector improved retinal sensitivity, albeit modestly and temporarily. Comparison with the results obtained in the dog model indicates that there is a species difference in the amount of RPE65 required to drive the visual cycle and that the demand for RPE65 in affected persons was not met to the extent required for a durable, robust effect. (Funded by the National Institute for Health Research and others; ClinicalTrials.gov number, NCT00643747.).


Subject(s)
DNA, Complementary/administration & dosage , Genetic Therapy , Genetic Vectors/administration & dosage , Leber Congenital Amaurosis/therapy , Retina/physiology , cis-trans-Isomerases/genetics , Adolescent , Animals , Child , Dependovirus , Disease Models, Animal , Disease Progression , Dogs , Humans , Leber Congenital Amaurosis/genetics , Mutation , Photoreceptor Cells, Vertebrate , Vision, Ocular , Young Adult
18.
Br Med Bull ; 126(1): 13-25, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29506236

ABSTRACT

Introduction: Inherited retinal diseases are the leading cause of sight impairment in people of working age in England and Wales, and the second commonest in childhood. Gene therapy offers the potential for benefit. Sources of data: Pubmed and clinicaltrials.gov. Areas of agreement: Gene therapy can improve vision in RPE65-associated Leber Congenital Amaurosis (RPE65-LCA). Potential benefit depends on efficient gene transfer and is limited by the extent of retinal degeneration. Areas of controversy: The magnitude of vision improvement from RPE65-LCA gene therapy is suboptimal, and its durability may be limited by progressive retinal degeneration. Growing points: The safety and potential benefit of gene therapy for inherited and acquired retinal diseases is being explored in a rapidly expanding number of trials. Areas timely for developing research: Developments in vector design and delivery will enable greater efficiency and safety of gene transfer. Optimization of trial design will accelerate reliable assessment of outcomes.


Subject(s)
Genetic Therapy/methods , Leber Congenital Amaurosis/therapy , Retinal Degeneration/genetics , Clinical Trials as Topic , Evidence-Based Medicine , Gene Transfer Techniques , Genetic Therapy/trends , Humans , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/physiopathology , Retinal Degeneration/physiopathology
19.
Ophthalmology ; 125(11): 1765-1775, 2018 11.
Article in English | MEDLINE | ID: mdl-29884405

ABSTRACT

PURPOSE: Transplantation of human embryonic stem cell (hESC)-derived retinal pigment epithelial (RPE) cells offers the potential for benefit in macular degeneration. Previous trials have reported improved visual acuity (VA), but lacked detailed analysis of retinal structure and function in the treated area. DESIGN: Phase 1/2 open-label dose-escalation trial to evaluate safety and potential efficacy (clinicaltrials.gov identifier, NCT01469832). PARTICIPANTS: Twelve participants with advanced Stargardt disease (STGD1), the most common cause of macular degeneration in children and young adults. METHODS: Subretinal transplantation of up to 200 000 hESC-derived RPE cells with systemic immunosuppressive therapy for 13 weeks. MAIN OUTCOME MEASURES: The primary end points were the safety and tolerability of hESC-derived RPE cell administration. We also investigated evidence of the survival of transplanted cells and measured retinal structure and function using microperimetry and spectral-domain OCT. RESULTS: Focal areas of subretinal hyperpigmentation developed in all participants in a dose-dependent manner in the recipient retina and persisted after withdrawal of systemic immunosuppression. We found no evidence of uncontrolled proliferation or inflammatory responses. Borderline improvements in best-corrected VA in 4 participants either were unsustained or were matched by a similar improvement in the untreated contralateral eye. Microperimetry demonstrated no evidence of benefit at 12 months in the 12 participants. In one instance at the highest dose, localized retinal thinning and reduced sensitivity in the area of hyperpigmentation suggested the potential for harm. Participant-reported quality of life using the 25-item National Eye Institute Visual Function Questionnaire indicated no significant change. CONCLUSIONS: Subretinal hyperpigmentation is consistent with the survival of viable transplanted hESC-derived RPE cells, but may reflect released pigment in their absence. The findings demonstrate the value of detailed analysis of spatial correlation of retinal structure and function in determining with appropriate sensitivity the impact of cell transplantation and suggest that intervention in early stage of disease should be approached with caution. Given the slow rate of progressive degeneration at this advanced stage of disease, any protection against further deterioration may be evident only after a more extended period of observation.


Subject(s)
Human Embryonic Stem Cells/transplantation , Macular Degeneration/congenital , Retinal Pigment Epithelium/transplantation , Adult , Electroretinography , Female , Fluorescein Angiography , Humans , Immunosuppressive Agents/therapeutic use , Macular Degeneration/diagnostic imaging , Macular Degeneration/physiopathology , Macular Degeneration/therapy , Male , Middle Aged , Photoreceptor Cells, Vertebrate/physiology , Quality of Life , Sickness Impact Profile , Slit Lamp Microscopy , Stargardt Disease , Tomography, Optical Coherence , Visual Acuity/physiology , Visual Field Tests , Visual Fields/physiology
20.
Adv Exp Med Biol ; 1074: 91-99, 2018.
Article in English | MEDLINE | ID: mdl-29721932

ABSTRACT

Neuronal ceroid lipofuscinoses (NCLs) are a group of fatal, inherited lysosomal storage disorders mostly affecting the central nervous system of children. Symptoms include vision loss, seizures, motor deterioration and cognitive decline ultimately resulting in premature death. Studies in animal models showed that the diseases are amenable to gene supplementation therapies, and over the last decade, major advances have been made in the (pre)clinical development of these therapies. This mini-review summarises and discusses current gene therapy approaches for NCL targeting the brain and the eye.


Subject(s)
Genetic Therapy/methods , Nerve Degeneration/therapy , Neuronal Ceroid-Lipofuscinoses/therapy , Vision Disorders/therapy , Animals , Brain/enzymology , Child , Clinical Trials as Topic , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/administration & dosage , Genetic Vectors/therapeutic use , Humans , Infant , Injections, Intraocular , Injections, Intraventricular , Lysosomes/enzymology , Neuronal Ceroid-Lipofuscinoses/complications , Neuronal Ceroid-Lipofuscinoses/enzymology , Organ Specificity , Vision Disorders/etiology
SELECTION OF CITATIONS
SEARCH DETAIL