Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters

Publication year range
1.
Brain ; 147(6): 2085-2097, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38735647

ABSTRACT

Biallelic pathogenic variants in the PNPLA6 gene cause a broad spectrum of disorders leading to gait disturbance, visual impairment, anterior hypopituitarism and hair anomalies. PNPLA6 encodes neuropathy target esterase (NTE), yet the role of NTE dysfunction on affected tissues in the large spectrum of associated disease remains unclear. We present a systematic evidence-based review of a novel cohort of 23 new patients along with 95 reported individuals with PNPLA6 variants that implicate missense variants as a driver of disease pathogenesis. Measuring esterase activity of 46 disease-associated and 20 common variants observed across PNPLA6-associated clinical diagnoses unambiguously reclassified 36 variants as pathogenic and 10 variants as likely pathogenic, establishing a robust functional assay for classifying PNPLA6 variants of unknown significance. Estimating the overall NTE activity of affected individuals revealed a striking inverse relationship between NTE activity and the presence of retinopathy and endocrinopathy. This phenomenon was recaptured in vivo in an allelic mouse series, where a similar NTE threshold for retinopathy exists. Thus, PNPLA6 disorders, previously considered allelic, are a continuous spectrum of pleiotropic phenotypes defined by an NTE genotype:activity:phenotype relationship. This relationship, and the generation of a preclinical animal model, pave the way for therapeutic trials, using NTE as a biomarker.


Subject(s)
Phenotype , Animals , Female , Humans , Male , Mice , Acyltransferases , Carboxylic Ester Hydrolases/genetics , Mutation, Missense , Phospholipases/genetics , Retinal Diseases/genetics
2.
Proc Natl Acad Sci U S A ; 117(9): 5016-5027, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32071204

ABSTRACT

During development, neural progenitors change their competence states over time to sequentially generate different types of neurons and glia. Several cascades of temporal transcription factors (tTFs) have been discovered in Drosophila to control the temporal identity of neuroblasts, but the temporal regulation mechanism is poorly understood in vertebrates. Mammalian retinal progenitor cells (RPCs) give rise to several types of neuronal and glial cells following a sequential yet overlapping temporal order. Here, by temporal cluster analysis, RNA-sequencing analysis, and loss-of-function and gain-of-function studies, we show that the Fox domain TF Foxn4 functions as a tTF during retinogenesis to confer RPCs with the competence to generate the mid/late-early cell types: amacrine, horizontal, cone, and rod cells, while suppressing the competence of generating the immediate-early cell type: retinal ganglion cells (RGCs). In early embryonic retinas, Foxn4 inactivation causes down-regulation of photoreceptor marker genes and decreased photoreceptor generation but increased RGC production, whereas its overexpression has the opposite effect. Just as in Drosophila, Foxn4 appears to positively regulate its downstream tTF Casz1 while negatively regulating its upstream tTF Ikzf1. Moreover, retina-specific ablation of Foxn4 reveals that it may be indirectly involved in the synaptogenesis, establishment of laminar structure, visual signal transmission, and long-term maintenance of the retina. Together, our data provide evidence that Foxn4 acts as a tTF to bias RPCs toward the mid/late-early cell fates and identify a missing member of the tTF cascade that controls RPC temporal identities to ensure the generation of proper neuronal diversity in the retina.


Subject(s)
Eye Proteins/metabolism , Forkhead Transcription Factors/metabolism , Neurogenesis/physiology , Retina/metabolism , Animals , DNA-Binding Proteins , Drosophila , Eye Proteins/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Ikaros Transcription Factor , Mice , Mice, Knockout, ApoE , Neuroglia/cytology , Neuroglia/metabolism , RNA-Seq , Retina/cytology , Retinal Cone Photoreceptor Cells/classification , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Sequence Analysis , Transcription Factors
3.
PLoS Genet ; 16(12): e1009259, 2020 12.
Article in English | MEDLINE | ID: mdl-33362196

ABSTRACT

Rab-GTPases and associated effectors mediate cargo transport through the endomembrane system of eukaryotic cells, regulating key processes such as membrane turnover, signal transduction, protein recycling and degradation. Using developmental transcriptome data, we identified Rabgef1 (encoding the protein RabGEF1 or Rabex-5) as the only gene associated with Rab GTPases that exhibited strong concordance with retinal photoreceptor differentiation. Loss of Rabgef1 in mice (Rabgef1-/-) resulted in defects specifically of photoreceptor morphology and almost complete loss of both rod and cone function as early as eye opening; however, aberrant outer segment formation could only partly account for visual function deficits. RabGEF1 protein in retinal photoreceptors interacts with Rabaptin-5, and RabGEF1 absence leads to reduction of early endosomes consistent with studies in other mammalian cells and tissues. Electron microscopy analyses reveal abnormal accumulation of macromolecular aggregates in autophagosome-like vacuoles and enhanced immunostaining for LC3A/B and p62 in Rabgef1-/- photoreceptors, consistent with compromised autophagy. Transcriptome analysis of the developing Rabgef1-/- retina reveals altered expression of 2469 genes related to multiple pathways including phototransduction, mitochondria, oxidative stress and endocytosis, suggesting an early trajectory of photoreceptor cell death. Our results implicate an essential role of the RabGEF1-modulated endocytic and autophagic pathways in photoreceptor differentiation and homeostasis. We propose that RabGEF1 and associated components are potential candidates for syndromic traits that include a retinopathy phenotype.


Subject(s)
Autophagy , Endocytosis , Guanine Nucleotide Exchange Factors/genetics , Neurogenesis , Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Animals , Female , Guanine Nucleotide Exchange Factors/metabolism , Male , Mice , Mice, Inbred BALB C , Photoreceptor Cells/cytology , Retinal Degeneration/genetics , Transcriptome
4.
Int J Mol Sci ; 24(6)2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36982411

ABSTRACT

It is extremely difficult to achieve functional recovery after axonal injury in the adult central nervous system. The activation of G-protein coupled receptor 110 (GPR110, ADGRF1) has been shown to stimulate neurite extension in developing neurons and after axonal injury in adult mice. Here, we demonstrate that GPR110 activation partially restores visual function impaired by optic nerve injury in adult mice. Intravitreal injection of GPR110 ligands, synaptamide and its stable analogue dimethylsynaptamide (A8) after optic nerve crush significantly reduced axonal degeneration and improved axonal integrity and visual function in wild-type but not gpr110 knockout mice. The retina obtained from the injured mice treated with GPR110 ligands also showed a significant reduction in the crush-induced loss of retinal ganglion cells. Our data suggest that targeting GPR110 may be a viable strategy for functional recovery after optic nerve injury.


Subject(s)
Optic Nerve Injuries , Animals , Mice , Axons , Ligands , Mice, Knockout , Nerve Crush , Nerve Regeneration/physiology , Receptors, G-Protein-Coupled/genetics , Retina , Retinal Ganglion Cells/physiology
5.
Gene Ther ; 29(7-8): 431-440, 2022 08.
Article in English | MEDLINE | ID: mdl-34548657

ABSTRACT

Animal models of X-linked juvenile retinoschisis (XLRS) are valuable tools for understanding basic biochemical function of retinoschisin (RS1) protein and to investigate outcomes of preclinical efficacy and toxicity studies. In order to work with an eye larger than mouse, we generated and characterized an Rs1h-/y knockout rat model created by removing exon 3. This rat model expresses no normal RS1 protein. The model shares features of an early onset and more severe phenotype of human XLRS. The morphologic pathology includes schisis cavities at postnatal day 15 (p15), photoreceptors that are misplaced into the subretinal space and OPL, and a reduction of photoreceptor cell numbers by p21. By 6 mo age only 1-3 rows of photoreceptors nuclei remain, and the inner/outer segment layers and the OPL shows major changes. Electroretinogram recordings show functional loss with considerable reduction of both the a-wave and b-wave by p28, indicating early age loss and dysfunction of photoreceptors. The ratio of b-/a-wave amplitudes indicates impaired synaptic transmission to bipolar cells in addition. Supplementing the Rs1h-/y exon3-del retina with normal human RS1 protein using AAV8-RS1 delivery improved the retinal structure. This Rs1h-/y rat model provides a further tool to explore underlying mechanisms of XLRS pathology and to evaluate therapeutic intervention for the XLRS condition.


Subject(s)
Cell Adhesion Molecules , Eye Proteins , Retinoschisis , Animals , Cell Adhesion Molecules/genetics , Dietary Supplements , Electroretinography , Exons/genetics , Eye Proteins/genetics , Eye Proteins/metabolism , Humans , Phenotype , Rats , Retina/metabolism , Retinoschisis/genetics , Retinoschisis/pathology , Retinoschisis/therapy
6.
Int J Mol Sci ; 23(21)2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36361898

ABSTRACT

Bietti crystalline corneo-retinal dystrophy (BCD) is an autosomal recessive inherited retinal dystrophy characterized by multiple shimmering yellow-white deposits in the posterior pole of the retina in association with atrophy of the retinal pigment epithelium (RPE), pigment clumps, and choroidal atrophy and sclerosis. Blindness and severe visual damage are common in late-stage BCD patients. We generated a Cyp4v3 knockout mouse model to investigate the pathogenesis of BCD. This model exhibits decreased RPE numbers and signs of inflammation response in the retina. Rod photoreceptors were vulnerable to light-induced injury, showing increased deposits through fundoscopy, a decrease in thickness and a loss of cells in the ONL, and the degeneration of rod photoreceptors. These results suggest that an inflammatory response might be an integral part of the pathophysiology of BCD, suggesting that it might be reasonable for BCD patients to avoid strong light, and the results provide a useful model for evaluating the effects of therapeutic approaches.


Subject(s)
Retinal Diseases , Retinal Dystrophies , Mice , Animals , Cytochrome P450 Family 4/genetics , Mutation , Retinal Diseases/pathology , Disease Models, Animal , Atrophy
7.
Hum Mol Genet ; 28(20): 3355-3368, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31332443

ABSTRACT

MicroRNA-204 (miR-204) is expressed in pulmonary, renal, mammary and eye tissue, and its reduction can result in multiple diseases including cancer. We first generated miR-204-/- mice to study the impact of miR-204 loss on retinal and retinal pigment epithelium (RPE) structure and function. The RPE is fundamentally important for maintaining the health and integrity of the retinal photoreceptors. miR-204-/- eyes evidenced areas of hyper-autofluorescence and defective photoreceptor digestion, along with increased microglia migration to the RPE. Migratory Iba1+ microglial cells were localized to the RPE apical surface where they participated in the phagocytosis of photoreceptor outer segments (POSs) and contributed to a persistent build-up of rhodopsin. These structural, molecular and cellular outcomes were accompanied by decreased light-evoked electrical responses from the retina and RPE. In parallel experiments, we suppressed miR-204 expression in primary cultures of human RPE using anti-miR-204. In vitro suppression of miR-204 in human RPE similarly showed abnormal POS clearance and altered expression of autophagy-related proteins and Rab22a, a regulator of endosome maturation. Together, these in vitro and in vivo experiments suggest that the normally high levels of miR-204 in RPE can mitigate disease onset by preventing generation of oxidative stress and inflammation originating from intracellular accumulation of undigested photoreactive POS lipids. More generally, these results implicate RPE miR-204-mediated regulation of autophagy and endolysosomal interaction as a critical determinant of normal RPE/retina structure and function.


Subject(s)
MicroRNAs/metabolism , Phagocytosis/physiology , Phagosomes/metabolism , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Electrophysiology , Female , Flow Cytometry , Humans , Lysosomal-Associated Membrane Protein 2/metabolism , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Phagocytosis/genetics , Phagosomes/physiology , Retina/physiology , Retinal Pigment Epithelium/physiology , Reverse Transcriptase Polymerase Chain Reaction
8.
Proc Natl Acad Sci U S A ; 114(21): E4271-E4280, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28484004

ABSTRACT

The polycistronic miR-183/96/182 cluster is preferentially and abundantly expressed in terminally differentiating sensory epithelia. To clarify its roles in the terminal differentiation of sensory receptors in vivo, we deleted the entire gene cluster in mouse germline through homologous recombination. The miR-183/96/182 null mice display impairment of the visual, auditory, vestibular, and olfactory systems, attributable to profound defects in sensory receptor terminal differentiation. Maturation of sensory receptor precursors is delayed, and they never attain a fully differentiated state. In the retina, delay in up-regulation of key photoreceptor genes underlies delayed outer segment elongation and possibly mispositioning of cone nuclei in the retina. Incomplete maturation of photoreceptors is followed shortly afterward by early-onset degeneration. Cell biologic and transcriptome analyses implicate dysregulation of ciliogenesis, nuclear translocation, and an epigenetic mechanism that may control timing of terminal differentiation in developing photoreceptors. In both the organ of Corti and the vestibular organ, impaired terminal differentiation manifests as immature stereocilia and kinocilia on the apical surface of hair cells. Our study thus establishes a dedicated role of the miR-183/96/182 cluster in driving the terminal differentiation of multiple sensory receptor cells.


Subject(s)
Hair Cells, Auditory/cytology , Hair Cells, Vestibular/cytology , MicroRNAs/genetics , Olfactory Mucosa/cytology , Retinal Cone Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/cytology , Animals , Gene Expression Regulation, Developmental/genetics , Hair Cells, Auditory/metabolism , Hair Cells, Vestibular/metabolism , Hearing Disorders/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Multigene Family , Olfaction Disorders/genetics , Olfactory Mucosa/metabolism , Postural Balance/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Sensation Disorders/genetics , Vision Disorders/genetics
9.
Hum Mutat ; 40(4): 426-443, 2019 04.
Article in English | MEDLINE | ID: mdl-30628748

ABSTRACT

Human RPE65 mutations cause a spectrum of retinal dystrophies that result in blindness. While RPE65 mutations have been almost invariably recessively inherited, a c.1430A>G (p.(D477G)) mutation has been reported to cause autosomal dominant retinitis pigmentosa (adRP). To study the pathogenesis of this human mutation, we have replicated the mutation in a knock-in (KI) mouse model using CRISPR/Cas9-mediated genome editing. Significantly, in contrast to human patients, heterozygous KI mice do not exhibit any phenotypes in visual function tests. When raised in regular vivarium conditions, homozygous KI mice display relatively undisturbed visual functions with minimal retinal structural changes. However, KI/KI mouse retinae are more sensitive to light exposure and exhibit signs of degenerative features when subjected to light stress. We find that instead of merely producing a missense mutant protein, the A>G nucleotide substitution greatly affects appropriate splicing of Rpe65 mRNA by generating an ectopic splice site in comparable context to the canonical one, thereby disrupting RPE65 protein expression. Similar splicing defects were also confirmed for the human RPE65 c.1430G mutant in an in vitro Exontrap assay. Our data demonstrate that a splicing defect is associated with c.1430G pathogenesis, and therefore provide insights in the therapeutic strategy for human patients.


Subject(s)
Alleles , Genetic Predisposition to Disease , Mutation , RNA Splicing , cis-trans-Isomerases/genetics , Animals , Biomarkers , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Genetic Association Studies , Genotype , Humans , Mice , Mice, Transgenic , Phenotype , RNA Splice Sites , Retina/metabolism , Retina/pathology
10.
Gene Ther ; 26(9): 386-398, 2019 09.
Article in English | MEDLINE | ID: mdl-31308478

ABSTRACT

Recombinant adeno-associated virus (rAAV) has been widely used for gene delivery in animal models and successfully applied in clinical trials for treating inherited retinal disease. Although subretinal delivery of AAVs can effectively transduce photoreceptors and/or retinal pigmental epithelium (RPE), cells most affected by inherited retinal diseases, the procedure is invasive and complicated, and only delivers the gene to a limited retinal area. AAVs can also be delivered intravitreally to the retina, a much less invasive nonsurgical procedure. However, intravitreal administration of non-modified AAV serotypes tends to transduce only ganglion cells and inner nuclear layer cells. To date, most non-modified AAV serotypes that have been identified are incapable of efficiently transducing photoreceptors and/or RPE when delivered intravitreally. In this study, we investigate the retinal tropism of AAVrh10 vector administered by intravitreal injection to mouse, rat, and rabbit eyes. Our results demonstrate that AAVrh10 is capable of transducing not only inner retinal cells, but also outer retinal cells in all three species, though the transduction efficiency in rabbit was low. In addition, AAVrh10 preferentially transduced outer retinal cells in mouse models of retinal disease. Therefore, AAVrh10 vector could be a useful candidate to intravitreally deliver genes to photoreceptor and RPE cells.


Subject(s)
Dependovirus/genetics , Retina , Transduction, Genetic/methods , Animals , Cytomegalovirus/genetics , Dependovirus/physiology , Genetic Vectors , Green Fluorescent Proteins/genetics , Intravitreal Injections , Male , Mice , Photoreceptor Cells/virology , Rabbits , Rats , Rats, Sprague-Dawley , Retina/drug effects , Retina/virology , Retinal Diseases/therapy , Viral Tropism
11.
Hum Mol Genet ; 26(12): 2218-2230, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28369466

ABSTRACT

In retinal photoreceptors, vectorial transport of cargo is critical for transduction of visual signals, and defects in intracellular trafficking can lead to photoreceptor degeneration and vision impairment. Molecular signatures associated with routing of transport vesicles in photoreceptors are poorly understood. We previously reported the identification of a novel rod photoreceptor specific isoform of Receptor Expression Enhancing Protein (REEP) 6, which belongs to a family of proteins involved in intracellular transport of receptors to the plasma membrane. Here we show that loss of REEP6 in mice (Reep6-/-) results in progressive retinal degeneration. Rod photoreceptor dysfunction is observed in Reep6-/- mice as early as one month of age and associated with aberrant accumulation of vacuole-like structures at the apical inner segment and reduction in selected rod phototransduction proteins. We demonstrate that REEP6 is detected in a subset of Clathrin-coated vesicles and interacts with the t-SNARE, Syntaxin3. In concordance with the rod degeneration phenotype in Reep6-/- mice, whole exome sequencing identified homozygous REEP6-E75K mutation in two retinitis pigmentosa families of different ethnicities. Our studies suggest a critical function of REEP6 in trafficking of cargo via a subset of Clathrin-coated vesicles to selected membrane sites in retinal rod photoreceptors.


Subject(s)
Membrane Transport Proteins/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Clathrin-Coated Vesicles/metabolism , Eye Proteins/genetics , Light Signal Transduction , Membrane Proteins , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Mutation , Photoreceptor Cells, Vertebrate/metabolism , Protein Isoforms/metabolism , Protein Transport , Qa-SNARE Proteins/metabolism , Retinal Degeneration/metabolism , Retinitis Pigmentosa/genetics , SNARE Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 113(21): E2925-34, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27162334

ABSTRACT

Mutations in the X-linked retinitis pigmentosa GTPase regulator (RPGR) gene are a major cause of retinitis pigmentosa, a blinding retinal disease resulting from photoreceptor degeneration. A photoreceptor specific ORF15 variant of RPGR (RPGR(ORF15)), carrying multiple Glu-Gly tandem repeats and a C-terminal basic domain of unknown function, localizes to the connecting cilium where it is thought to regulate cargo trafficking. Here we show that tubulin tyrosine ligase like-5 (TTLL5) glutamylates RPGR(ORF15) in its Glu-Gly-rich repetitive region containing motifs homologous to the α-tubulin C-terminal tail. The RPGR(ORF15) C-terminal basic domain binds to the noncatalytic cofactor interaction domain unique to TTLL5 among TTLL family glutamylases and targets TTLL5 to glutamylate RPGR. Only TTLL5 and not other TTLL family glutamylases interacts with RPGR(ORF15) when expressed transiently in cells. Consistent with this, a Ttll5 mutant mouse displays a complete loss of RPGR glutamylation without marked changes in tubulin glutamylation levels. The Ttll5 mutant mouse develops slow photoreceptor degeneration with early mislocalization of cone opsins, features resembling those of Rpgr-null mice. Moreover TTLL5 disease mutants that cause human retinal dystrophy show impaired glutamylation of RPGR(ORF15) Thus, RPGR(ORF15) is a novel glutamylation substrate, and this posttranslational modification is critical for its function in photoreceptors. Our study uncovers the pathogenic mechanism whereby absence of RPGR(ORF15) glutamylation leads to retinal pathology in patients with TTLL5 gene mutations and connects these two genes into a common disease pathway.


Subject(s)
Carrier Proteins/metabolism , Eye Proteins/metabolism , Mutation , Opsins/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinitis Pigmentosa/metabolism , Animals , Carrier Proteins/genetics , Eye Proteins/genetics , Humans , Mice , Mice, Knockout , Opsins/genetics , Protein Domains , Retinal Cone Photoreceptor Cells/pathology , Retinitis Pigmentosa/genetics
13.
J Neurosci ; 37(12): 3294-3310, 2017 03 22.
Article in English | MEDLINE | ID: mdl-28235894

ABSTRACT

Photoreceptor degeneration is a cause of irreversible vision loss in incurable blinding retinal diseases including retinitis pigmentosa (RP) and atrophic age-related macular degeneration. We found in two separate mouse models of photoreceptor degeneration that tamoxifen, a selective estrogen receptor modulator and a drug previously linked with retinal toxicity, paradoxically provided potent neuroprotective effects. In a light-induced degeneration model, tamoxifen prevented onset of photoreceptor apoptosis and atrophy and maintained near-normal levels of electroretinographic responses. Rescue effects were correlated with decreased microglial activation and inflammatory cytokine production in the retina in vivo and a reduction of microglia-mediated toxicity to photoreceptors in vitro, indicating a microglia-mediated mechanism of rescue. Tamoxifen also rescued degeneration in a genetic (Pde6brd10) model of RP, significantly improving retinal structure, electrophysiological responses, and visual behavior. These prominent neuroprotective effects warrant the consideration of tamoxifen as a drug suitable for being repurposed to treat photoreceptor degenerative disease.SIGNIFICANCE STATEMENT Photoreceptor degeneration is a cause of irreversible blindness in a number of retinal diseases such as retinitis pigmentosa (RP) and atrophic age-related macular degeneration. Tamoxifen, a selective estrogen receptor modulator approved for the treatment of breast cancer and previously linked to a low incidence of retinal toxicity, was unexpectedly found to exert marked protective effects against photoreceptor degeneration. Structural and functional protective effects were found for an acute model of light-induced photoreceptor injury and for a genetic model for RP. The mechanism of protection involved the modulation of microglial activation and the production of inflammatory cytokines, highlighting the role of inflammatory mechanisms in photoreceptor degeneration. Tamoxifen may be suitable for clinical study as a potential treatment for diseases involving photoreceptor degeneration.


Subject(s)
Nerve Regeneration/physiology , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/drug therapy , Retinal Degeneration/physiopathology , Tamoxifen/administration & dosage , Animals , Apoptosis/drug effects , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Inbred C57BL , Nerve Regeneration/radiation effects , Neuroprotective Agents/administration & dosage , Photoreceptor Cells, Vertebrate/physiology , Recovery of Function/drug effects , Retinal Degeneration/pathology , Treatment Outcome
14.
Hum Mol Genet ; 24(22): 6446-58, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26358772

ABSTRACT

Retinal neurodegenerative diseases are especially attractive targets for gene replacement therapy, which appears to be clinically effective for several monogenic diseases. X-linked forms of retinitis pigmentosa (XLRP) are relatively severe blinding disorders, resulting from progressive photoreceptor dysfunction primarily caused by mutations in RPGR or RP2 gene. With a goal to develop gene therapy for the XLRP-RP2 disease, we first performed detailed characterization of the Rp2-knockout (Rp2-KO) mice and observed early-onset cone dysfunction, which was followed by progressive cone degeneration, mimicking cone vision impairment in XLRP patients. The mice also exhibited distinct and significantly delayed falling phase of photopic b-wave of electroretinogram (ERG). Concurrently, we generated a self-complementary adeno-associated viral (AAV) vector carrying human RP2-coding sequence and demonstrated its ability to mediate stable RP2 protein expression in mouse photoreceptors. A long-term efficacy study was then conducted in Rp2-KO mice following AAV-RP2 vector administration. Preservation of cone function was achieved with a wide dose range over 18-month duration, as evidenced by photopic ERG and optomotor tests. The slower b-wave kinetics was also completely restored. Morphologically, the treatment preserved cone viability, corrected mis-trafficking of M-cone opsin and restored cone PDE6 expression. The therapeutic effect was achieved even in mice that received treatment at an advanced disease stage. The highest AAV-RP2 dose group demonstrated retinal toxicity, highlighting the importance of careful vector dosing in designing future human trials. The wide range of effective dose, a broad treatment window and long-lasting therapeutic effects should make the RP2 gene therapy attractive for clinical development.


Subject(s)
Eye Proteins/genetics , Genetic Therapy/methods , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Retinal Cone Photoreceptor Cells/physiology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Animals , Electroretinography , Eye Proteins/biosynthesis , GTP-Binding Proteins , Genetic Diseases, X-Linked/genetics , Genetic Vectors , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Membrane Proteins/biosynthesis , Mice , Mice, Knockout , Mutation , Pyrophosphatases/deficiency , Pyrophosphatases/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/genetics , Retinitis Pigmentosa/metabolism
15.
Hum Mol Genet ; 24(14): 3956-70, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25877300

ABSTRACT

Mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene account for >70% of X-linked retinitis pigmentosa (XLRP) and 15-20% of all inherited retinal degeneration. Gene replacement therapy for RPGR-XLRP was hampered by the relatively slow disease progression in mouse models and by difficulties in cloning the full-length RPGR-ORF15 cDNA that includes a purine-rich 3'-coding region; however, its effectiveness has recently been demonstrated in four dogs with RPGR mutations. To advance the therapy to clinical stage, we generated new stable vectors in AAV8 or AAV9 carrying mouse and human full-length RPGR-ORF15-coding sequence and conducted a comprehensive long-term dose-efficacy study in Rpgr-knockout mice. After validating their ability to produce full-length proteins that localize to photoreceptor connecting cilia, we evaluated various vector doses in mice during a 2-year study. We demonstrate that eyes treated with a single injection of mouse or human RPGR-ORF15 vector at an optimal dose maintained the expression of RPGR-ORF15 throughout the study duration and exhibited higher electroretinogram amplitude, thicker photoreceptor layer and better targeting of opsins to outer segments compared with sham-treated eyes. Furthermore, mice that received treatment at an advanced age also showed remarkable preservation of retinal structure and function. Retinal toxicity was observed at high vector doses, highlighting the importance of careful dose optimization in future clinical experiments. Our long-term dose-efficacy study should facilitate the design of human trials with human RPGR-ORF15 vector as a clinical candidate.


Subject(s)
Carrier Proteins/genetics , Eye Proteins/genetics , Genetic Therapy , Retinitis Pigmentosa/genetics , Animals , Carrier Proteins/metabolism , Dependovirus/genetics , Disease Models, Animal , Electroretinography , Exons , Eye Proteins/metabolism , Genetic Vectors/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Open Reading Frames , Retina/metabolism , Retinitis Pigmentosa/metabolism
16.
Hum Mol Genet ; 24(15): 4417-28, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25972377

ABSTRACT

Human RPE65 mutations cause a spectrum of blinding retinal dystrophies from severe early-onset disease to milder manifestations. The RPE65 P25L missense mutation, though having <10% of wild-type (WT) activity, causes relatively mild retinal degeneration. To better understand these mild forms of RPE65-related retinal degeneration, and their effect on cone photoreceptor survival, we generated an Rpe65/P25L knock-in (KI/KI) mouse model. We found that, when subject to the low-light regime (∼100 lux) of regular mouse housing, homozygous Rpe65/P25L KI/KI mice are morphologically and functionally very similar to WT siblings. While mutant protein expression is decreased by over 80%, KI/KI mice retinae retain comparable 11-cis-retinal levels with WT. Consistently, the scotopic and photopic electroretinographic (ERG) responses to single-flash stimuli also show no difference between KI/KI and WT mice. However, the recovery of a-wave response following moderate visual pigment bleach is delayed in KI/KI mice. Importantly, KI/KI mice show significantly increased resistance to high-intensity (20 000 lux for 30 min) light-induced retinal damage (LIRD) as compared with WT, indicating impaired rhodopsin regeneration in KI/KI. Taken together, the Rpe65/P25L mutant produces sufficient chromophore under normal conditions to keep opsins replete and thus manifests a minimal phenotype. Only when exposed to intensive light is this hypomorphic mutation manifested physiologically, as its reduced expression and catalytic activity protects against the successive cycles of opsin regeneration underlying LIRD. These data also help define minimal requirements of chromophore for photoreceptor survival in vivo and may be useful in assessing a beneficial therapeutic dose for RPE65 gene therapy in humans.


Subject(s)
Retina/metabolism , Retinal Degeneration/genetics , Retinaldehyde/genetics , cis-trans-Isomerases/genetics , Animals , Disease Models, Animal , Gene Knock-In Techniques , Humans , Light , Mice , Mutation, Missense , Opsins/genetics , Opsins/metabolism , Retina/pathology , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/physiopathology , Retinaldehyde/biosynthesis , cis-trans-Isomerases/metabolism
17.
J Transl Med ; 13: 330, 2015 Oct 17.
Article in English | MEDLINE | ID: mdl-26476672

ABSTRACT

BACKGROUND: The wingless-type MMTV integration site (Wnt) signaling is a group of signal transduction pathways. In canonical Wnt pathway, Wnt ligands bind to low-density lipoprotein receptor-related protein 5 or 6 (LRP5 or LRP6), resulting in phosphorylation and activation of the receptor. We hypothesize that canonical Wnt pathway plays a role in the retinal lesion of age-related macular degeneration (AMD), a leading cause of irreversible central visual loss in elderly. METHODS: We examined LRP6 phosphorylation and Wnt signaling cascade in human retinal sections and plasma kallistatin, an endogenous inhibitor of the Wnt pathway in AMD patients and non-AMD subjects. We also used the Ccl2 (-/-) /Cx3cr1 (-/-) /rd8 and Ccl2 (-/-) /Cx3cr1 (gfp/gfp) mouse models with AMD-like retinal degeneration to further explore the involvement of Wnt signaling activation in the retinal lesions in those models and to preclinically evaluate the role of Wnt signaling suppression as a potential therapeutic option for AMD. RESULTS: We found higher levels of LRP6 (a key Wnt signaling receptor) protein phosphorylation and transcripts of the Wnt pathway-targeted genes, as well as higher beta-catenin protein in AMD macula compared to controls. Kallistatin was decreased in the plasma of AMD patients. Retinal non-phosphorylated-ß-catenin and phosphorylated-LRP6 were higher in Ccl2 (-/-) /Cx3cr1 (-/-) /rd8 mice than that in wild type. Intravitreal administration of an anti-LRP6 antibody slowed the progression of retinal lesions in Ccl2 (-/-) /Cx3cr1 (-/-) /rd8 and Ccl2 (-/-) /Cx3cr1 (gfp/gfp) mice. Electroretinography of treated eyes exhibited larger amplitudes compared to controls in both mouse models. A2E, a retinoid byproduct associated with AMD was lower in the treated eyes of Ccl2 (-/-) /Cx3cr1 (-/-) /rd8 mice. Anti-LRP6 also suppressed the expression of Tnf-α and Icam-1 in Ccl2 (-/-) /Cx3cr1 (-/-) /rd8 retinas. CONCLUSIONS: Wnt signaling may be disturbed in AMD patients, which could contribute to the retinal inflammation and increased A2E levels found in AMD. Aberrant activation of canonical Wnt signaling might also contribute to the focal retinal degenerative lesions of mouse models with Ccl2 and Cx3cr1 deficiency, and intravitreal administration of anti-LRP6 antibody could be beneficial by deactivating the canonical Wnt pathway.


Subject(s)
Gene Expression Regulation , Macular Degeneration/blood , Wnt Proteins/metabolism , Aged , Aging , Animals , CX3C Chemokine Receptor 1 , Chemokine CCL2/genetics , Disease Models, Animal , Electroretinography , Female , Humans , Intravitreal Injections , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Macular Degeneration/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Phosphorylation , Receptors, Chemokine/genetics , Retina/metabolism , Retinal Degeneration , Serpins/blood , Signal Transduction
18.
Exp Eye Res ; 139: 48-63, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26164072

ABSTRACT

All three classes of receptors for the inhibitory neurotransmitter GABA (GABAR) are expressed in the retina. This study investigated roles of GABAR, especially GABACR (GABA(A)-ρ), in retinal signaling in vivo by studying effects on the mouse electroretinogram (ERG) of genetic deletion of GABACR versus pharmacological blockade using receptor antagonists. Brief full-field flash ERGs were recorded from anesthetized GABACR(-/-) mice, and WT C57BL/6 (B6) mice, before and after intravitreal injection of GABACR antagonists, TPMPA, 3-APMPA, or the more recently developed 2-AEMP; GABAAR antagonist, SR95531; GABABR antagonist, CGP, and agonist, baclofen. Intravitreal injections of TPMPA and SR95531 were also made in Brown Norway rats. The effect of 2-AEMP on GABA-induced current was tested directly in isolated rat rod bipolar cells, and 2-AEMP was found to preferentially block GABACR in those cells. Maximum amplitudes of dark (DA) and light-adapted (LA) ERG b-waves were reduced in GABACR(-/-) mice, compared to B6 mice, by 30-60%; a-waves were unaltered and oscillatory potential amplitudes were increased. In B6 mice, after injection of TPMPA (also in rats), 3-APMPA or 2-AEMP, ERGs became similar to ERGs of GABACR(-/-) mice. Blockade of GABAARs and GABABRs, or agonism of GABABRs did not alter B6 DA b-wave amplitude. The negative scotopic threshold response (nSTR) was slightly less sensitive in GABACR(-/-) than in B6 mice, and unaltered by 2-AEMP. However, amplitudes of nSTR and photopic negative response (PhNR), both of which originate from inner retina, were enhanced by TPMPA and 3-APMPA, each of which has GABAB agonist properties, and further increased by baclofen. The finding that genetic deletion of GABACR, the GABACR antagonist 2-AEMP, and other antagonists all reduced ERG b-wave amplitude, supports a role for GABACR in determining the maximum response amplitude of bipolar cells contributing to the b-wave. GABACR antagonists differed in their effects on nSTR and PhNR; antagonists with GABAB agonist properties enhanced light-driven responses whereas 2-AEMP did not.


Subject(s)
DNA/genetics , Electroretinography , Gene Expression Regulation , Receptors, GABA/genetics , Retina/metabolism , Retinal Diseases/genetics , Animals , Cells, Cultured , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptors, GABA/biosynthesis , Retina/pathology , Retina/physiopathology , Retinal Diseases/metabolism , Retinal Diseases/physiopathology
19.
Vis Neurosci ; 32: E004, 2015 Jan.
Article in English | MEDLINE | ID: mdl-26241901

ABSTRACT

Electroretinogram (ERG) studies identified a new mouse line with a normal a-wave but lacking the b-wave component. The ERG phenotype of this new allele, nob7, matched closely that of mouse mutants for Grm6, Lrit3, Trpm1, and Nyx, which encode for proteins expressed in depolarizing bipolar cells (DBCs). To identify the underlying mutation, we first crossed nob7 mice with Grm6 nob3 mutants and measured the ERGs in offspring. All the offspring lacked the b-wave, indicating that nob7 is a new allele for Grm6: Grm6 nob7 . Sequence analyses of Grm6 nob7 cDNAs identified a 28 base pair insertion between exons 8 and 9, which would result in a frameshift mutation in the open reading frame that encodes the metabotropic glutamate receptor 6 (Grm6). Sequencing both the cDNA and genomic DNA from exon 8 and intron 8, respectively, from the Grm6 nob7 mouse revealed a G to A transition at the last position in exon 8. This mutation disrupts splicing and the normal exon 8 is extended by 28 base pairs, because splicing occurs 28 base pairs downstream at a cryptic splice donor. Consistent with the impact of the resulting frameshift mutation, there is a loss of mGluR6 protein (encoded by Grm6) from the dendritic tips of DBCs in the Grm6 nob7 retina. These results indicate that Grm6 nob7 is a new model of the complete form of congenital stationary night blindness, a human condition that has been linked to mutations of GRM6.


Subject(s)
Eye Diseases, Hereditary/diagnosis , Eye Diseases, Hereditary/genetics , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/genetics , Mutation/genetics , Myopia/diagnosis , Myopia/genetics , Night Blindness/diagnosis , Night Blindness/genetics , Receptors, Metabotropic Glutamate/genetics , Retina/pathology , Animals , Calcium Channels, N-Type/metabolism , Dark Adaptation/genetics , Disease Models, Animal , Electroretinography , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , Retina/metabolism
20.
Adv Exp Med Biol ; 801: 395-9, 2014.
Article in English | MEDLINE | ID: mdl-24664723

ABSTRACT

LMAN1 is a type I transmembrane protein that selectively transports its cargo proteins from ER to ER-Golgi intermediate compartment (ERGIC) and Golgi. Lman1 is a direct target of the transcription factor NRL in mouse retina. Therefore, we examined the in vivo function of LMAN1 in retina. Although Lman1 (- / -) mouse eyes did not show abnormality in histology and electroretinogram analysis at 3 months, Lman1 (- / -) retina at 6 months showed a decrease in cis-Golgi markers GM130 and GRASP65. We also observed abnormal level and location of Rhodopsin in these mice. Taken together, LMAN1 may play a role in photoreceptor gene transport and homeostasis.


Subject(s)
Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Homeostasis/physiology , Mannose-Binding Lectins/metabolism , Membrane Proteins/metabolism , Photoreceptor Cells, Vertebrate/physiology , Animals , Biological Transport/physiology , Mannose-Binding Lectins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL