Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 36
1.
Int J Mol Sci ; 24(4)2023 Feb 14.
Article En | MEDLINE | ID: mdl-36835257

Age-related macular degeneration (AMD) is the most common cause of blindness in the aged population. However, to date there is no effective treatment for the dry form of the disease, representing 85-90% of cases. AMD is an immensely complex disease which affects, amongst others, both retinal pigment epithelium (RPE) and photoreceptor cells and leads to the progressive loss of central vision. Mitochondrial dysfunction in both RPE and photoreceptor cells is emerging as a key player in the disease. There are indications that during disease progression, the RPE is first impaired and RPE dysfunction in turn leads to subsequent photoreceptor cell degeneration; however, the exact sequence of events has not as yet been fully determined. We recently showed that AAV delivery of an optimised NADH-ubiquinone oxidoreductase (NDI1) gene, a nuclear-encoded complex 1 equivalent from S. cerevisiae, expressed from a general promoter, provided robust benefit in a variety of murine and cellular models of dry AMD; this was the first study employing a gene therapy to directly boost mitochondrial function, providing functional benefit in vivo. However, use of a restricted RPE-specific promoter to drive expression of the gene therapy enables exploration of the optimal target retinal cell type for dry AMD therapies. Furthermore, such restricted transgene expression could reduce potential off-target effects, possibly improving the safety profile of the therapy. Therefore, in the current study, we interrogate whether expression of the gene therapy from the RPE-specific promoter, Vitelliform macular dystrophy 2 (VMD2), might be sufficient to rescue dry AMD models.


Genetic Therapy , Geographic Atrophy , Saccharomyces cerevisiae Proteins , Aged , Animals , Humans , Mice , Electron Transport Complex I/metabolism , Genetic Therapy/methods , Geographic Atrophy/genetics , Geographic Atrophy/therapy , Mitochondria/metabolism , Retinal Pigment Epithelium/metabolism , Saccharomyces cerevisiae Proteins/genetics
3.
Mol Ther Methods Clin Dev ; 25: 236-249, 2022 Jun 09.
Article En | MEDLINE | ID: mdl-35474956

Recombinant adeno-associated virus (AAV) vectors are one of the main gene delivery vehicles used in retinal gene therapy approaches; however, there is a need to further improve the efficacy, tropism, and safety of these vectors. In this study, using a CMV-EGFP expression cassette, we characterize the retinal utility of AAV-PHP.eB, a serotype recently developed by in vivo directed evolution, which can cross the blood-brain barrier and target neurons with high efficacy in mice. Systemic and intravitreal delivery of AAV-PHP.eB resulted in the high transduction efficacy of retinal ganglion and horizontal cells, with systemic delivery providing pan-retinal coverage of the mouse retina. Subretinal delivery transduced photoreceptors and retinal pigment epithelium cells robustly. EGFP expression (number of transduced cells and mRNA levels) were similar when the retinas were transduced systemically or intravitreally with AAV-PHP.eB or intravitreally with AAV2/2. Notably, in photoreceptors, EGFP fluorescence intensities and mRNA levels were 50-70 times higher, when subretinal injections with AAV-PHP.eB were compared to AAV2/8. Our results demonstrate the pan-retinal transduction of ganglion cells and extremely efficient transduction of photoreceptor and retinal pigment epithelium cells as the most valuable features of AAV-PHP.eB in the mouse retina.

4.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article En | MEDLINE | ID: mdl-34502356

Previous studies have shown that glaucomatous Schlemm's canal endothelial cells (gSCECs) are stiffer and associated with reduced porosity and increased extracellular matrix (ECM) material compared to SCECs from healthy individuals. We hypothesised that Schlemm's canal (SC) cell stiffening was a function of fibrotic changes occurring at the inner wall of SC in glaucoma. This study was performed in primary cell cultures isolated from the SC lumen of human donor eyes. RNA and protein quantification of both fibrotic and endothelial cell markers was carried out on both healthy and gSCECs. Functional assays to assess cell density, size, migration, proliferation, and mitochondrial function of these cells were also carried out. Indeed, we found that gSCECs deviate from typical endothelial cell characteristics and exhibit a more fibrotic phenotype. For example, gSCECs expressed significantly higher protein levels of the fibrotic markers α-SMA, collagen I-α1, and fibronectin, as well as significantly increased protein expression of TGFß-2, the main driver of fibrosis, compared to healthy SCECs. Interestingly, we observed a significant increase in protein expression of endothelial marker VE-cadherin in gSCECs, compared to healthy SCECs. gSCECs also appeared to be significantly larger, and surprisingly proliferate and migrate at a significantly higher rate, as well as showing significantly reduced mitochondrial activity, compared to healthy SCECs.


Fibrosis/physiopathology , Glaucoma/metabolism , Glaucoma/physiopathology , Antigens, CD/metabolism , Aqueous Humor/metabolism , Cadherins/metabolism , Cell Count , Cell Movement , Cell Proliferation , Endothelial Cells/metabolism , Endothelium , Extracellular Matrix , Eye/metabolism , Humans , Mitochondria , Porosity , Primary Cell Culture , Sclera , Trabecular Meshwork , Transforming Growth Factor beta2/metabolism
5.
Int J Mol Sci ; 22(11)2021 May 26.
Article En | MEDLINE | ID: mdl-34073611

Inherited retinal diseases (IRDs) represent a collection of phenotypically and genetically diverse conditions. IRDs phenotype(s) can be isolated to the eye or can involve multiple tissues. These conditions are associated with diverse forms of inheritance, and variants within the same gene often can be associated with multiple distinct phenotypes. Such aspects of the IRDs highlight the difficulty met when establishing a genetic diagnosis in patients. Here we provide an overview of cutting-edge next-generation sequencing techniques and strategies currently in use to maximise the effectivity of IRD gene screening. These techniques have helped researchers globally to find elusive causes of IRDs, including copy number variants, structural variants, new IRD genes and deep intronic variants, among others. Resolving a genetic diagnosis with thorough testing enables a more accurate diagnosis and more informed prognosis and should also provide information on inheritance patterns which may be of particular interest to patients of a child-bearing age. Given that IRDs are heritable conditions, genetic counselling may be offered to help inform family planning, carrier testing and prenatal screening. Additionally, a verified genetic diagnosis may enable access to appropriate clinical trials or approved medications that may be available for the condition.


Genetic Counseling , Genetic Diseases, Inborn , Genetic Testing , High-Throughput Nucleotide Sequencing , Prenatal Diagnosis , Retinal Diseases , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Humans , Retinal Diseases/diagnosis , Retinal Diseases/genetics
6.
Mol Ther Methods Clin Dev ; 20: 86-94, 2021 Mar 12.
Article En | MEDLINE | ID: mdl-33376757

Systemic or localized application of glucocorticoids (GCs) can lead to iatrogenic ocular hypertension, which is a leading cause of secondary open-angle glaucoma and visual impairment. Previous work has shown that dexamethasone increases zonula occludens-1 (ZO-1) protein expression in trabecular meshwork (TM) cells, and that an antisense oligonucleotide inhibitor of ZO-1 can abolish the dexamethasone-induced increase in trans-endothelial flow resistance in cultured Schlemm's canal (SC) endothelial and TM cells. We have previously shown that intracameral inoculation of small interfering RNA (siRNA) targeting SC endothelial cell tight junction components, ZO-1 and tricellulin, increases aqueous humor outflow facility ex vivo in normotensive mice by reversibly opening SC endothelial paracellular pores. In this study, we show that targeted siRNA downregulation of these SC endothelial tight junctions reduces intraocular pressure (IOP) in vivo, with a concomitant increase in conventional outflow facility in a well-characterized chronic steroid-induced mouse model of ocular hypertension, thus representing a potential focused clinical application for this therapy in a sight-threatening scenario.

7.
Genes (Basel) ; 11(12)2020 11 27.
Article En | MEDLINE | ID: mdl-33261050

RPE65 isomerase, expressed in the retinal pigmented epithelium (RPE), is an enzymatic component of the retinoid cycle, converting all-trans retinyl ester into 11-cis retinol, and it is essential for vision, because it replenishes the photon capturing 11-cis retinal. To date, almost 200 loss-of-function mutations have been identified within the RPE65 gene causing inherited retinal dystrophies, most notably Leber congenital amaurosis (LCA) and autosomal recessive retinitis pigmentosa (arRP), which are both severe and early onset disease entities. We previously reported a mutation, D477G, co-segregating with the disease in a late-onset form of autosomal dominant RP (adRP) with choroidal involvement; uniquely, it is the only RPE65 variant to be described with a dominant component. Families or individuals with this variant have been encountered in five countries, and a number of subsequent studies have been reported in which the molecular biological and physiological properties of the variant have been studied in further detail, including observations of possible novel functions in addition to reduced RPE65 enzymatic activity. With regard to the latter, a human phase 1b proof-of-concept study has recently been reported in which aspects of remaining vision were improved for up to one year in four of five patients with advanced disease receiving a single one-week oral dose of 9-cis retinaldehyde, which is the first report showing efficacy and safety of an oral therapy for a dominant form of RP. Here, we review data accrued from published studies investigating molecular mechanisms of this unique variant and include hitherto unpublished material on the clinical spectrum of disease encountered in patients with the D477G variant, which, in many cases bears striking similarities to choroideremia.


Amino Acid Substitution , Genes, Dominant , Mutation, Missense , Point Mutation , Retinitis Pigmentosa/genetics , cis-trans-Isomerases/genetics , Age of Onset , Animals , Choroideremia , Clinical Trials, Phase I as Topic , DNA, Complementary/administration & dosage , DNA, Complementary/genetics , Enzyme Replacement Therapy , Female , Gene Knock-In Techniques , Genetic Therapy , Genetic Vectors/therapeutic use , Humans , Leber Congenital Amaurosis/enzymology , Leber Congenital Amaurosis/genetics , Male , Mice , Pedigree , Proof of Concept Study , Protein Isoforms/genetics , Retinaldehyde/therapeutic use , Retinitis Pigmentosa/diagnostic imaging , Retinitis Pigmentosa/enzymology , Retinitis Pigmentosa/therapy , cis-trans-Isomerases/deficiency , cis-trans-Isomerases/physiology , cis-trans-Isomerases/therapeutic use
8.
Front Neurosci ; 14: 891, 2020.
Article En | MEDLINE | ID: mdl-32973439

With marketing approval of the first ocular gene therapy, and other gene therapies in clinical trial, treatments for inherited retinal degenerations (IRDs) have become a reality. Biallelic mutations in the tubby like protein 1 gene (TULP1) are causative of IRDs in humans; a mouse knock-out model (Tulp1-/-) is characterized by a similar disease phenotype. We developed a Tulp1 supplementation therapy for Tulp1-/- mice. Utilizing subretinal AAV2/5 delivery at postnatal day (p)2-3 and rhodopsin-kinase promoter (GRK1P) we targeted Tulp1 to photoreceptor cells exploring three doses, 2.2E9, 3.7E8, and 1.2E8 vgs. Tulp1 mRNA and TULP1 protein were assessed by RT-qPCR, western blot and immunocytochemistry, and visual function by electroretinography. Our results indicate that TULP1 was expressed in photoreceptors; achieved levels of Tulp1 mRNA and protein were similar to wild type levels at p20. However, the thickness of the outer nuclear layer (ONL) did not improve in treated Tulp1-/- mice. There was a small and transient electroretinography benefit in the treated retinas at 4 weeks of age (not observed by 6 weeks) when using 3.7E8 vg dose. Dark-adapted mixed rod and cone a- and b-wave amplitudes were 24.3 ± 13.5 µV and 52.2 ± 31.7 µV in treated Tulp1-/- mice, which were significantly different (p < 0.001, t-test), from those detected in untreated eyes (7.1 ± 7.0 µV and 9.4 ± 15.1 µV, respectively). Our results indicate that Tulp1 supplementation in photoreceptors may not be sufficient to provide robust benefit in Tulp1-/- mice. As such, further studies are required to fine tune the Tulp1 supplementation therapy, which, in principle, should rescue the Tulp1-/- phenotype.

9.
Front Neurosci ; 14: 656, 2020.
Article En | MEDLINE | ID: mdl-32655363

Mutations in tubby like protein 1 gene (TULP1) are causative of early-onset recessive inherited retinal degenerations (IRDs); similarly, the Tulp1-/- mouse is also characterized by a rapid IRD. Tulp1 mRNA and protein expression was analyzed in wild type mouse retinas and expression data sets (NCBI) during early postnatal development. Comparative histology was undertaken in Tulp1-/-, rhodopsin-/- (Rho-/-) and retinal degeneration slow-/- (Rds-/-) mouse retinas. Bioinformatic analysis of predicted TULP1 interactors and IRD genes was performed. Peak expression of Tulp1 in healthy mouse retinas was detected at p8; of note, TULP1 was detected in both the outer and inner retina. Bioinformatic analysis indicated Tulp1 expression in retinal progenitor, photoreceptor and non-photoreceptor cells. While common features of photoreceptor degeneration were detected in Tulp1-/-, Rho-/-, and Rds-/- retinas, other alterations in bipolar, amacrine and ganglion cells were specific to Tulp1-/- mice. Additionally, predicted TULP1 interactors differed in various retinal cell types and new functions for TULP1 were suggested. A pilot bioinformatic analysis indicated that in a similar fashion to Tulp1, many other IRD genes were expressed in both inner and outer retinal cells at p4-p7. Our data indicate that expression of Tulp1 extends to multiple retinal cell types; lack of TULP1 may lead to primary degeneration not only of photoreceptor but also non-photoreceptor cells. Predicted interactors suggest widespread retinal functions for TULP1. Early and widespread expression of TULP1 and some other IRD genes in both the inner and outer retina highlights potential hurdles in the development of treatments for these IRDs.

10.
BMJ Open Ophthalmol ; 5(1): e000462, 2020.
Article En | MEDLINE | ID: mdl-32426524

OBJECTIVES: No therapeutic interventions are currently available for autosomal dominant retinitis pigmentosa (adRP). An RPE65 Asp477Gly transition associates with late-onset adRP, reduced RPE65 enzymatic activity being one feature associated with this dominant variant. Our objective: to assess whether in a proof-of-concept study, oral synthetic 9 cis-retinyl acetate therapy improves vision in such advanced disease. METHODS AND ANALYSIS: A phase 1b proof-of-concept clinical trial was conducted involving five patients with advanced disease, aged 41-68 years. Goldmann visual fields (GVF) and visual acuities (VA) were assessed for 6-12 months after 7-day treatment, patients receiving consecutive oral doses (40 mg/m2) of 9-cis-retinyl acetate, a synthetic retinoid replacement. RESULTS: Pathological effects of D477G variant were preliminarily assessed by electroretinography in mice expressing AAV-delivered D477G RPE65, by MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxyme- thoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] assays on RPE viability and enzyme activity in cultured cells. In addition to a mild dominant effect reflected in reduced electroretinographics in mice, and reduced cellular function in vitro, D477G exhibited reduced enzymatic RPE65 activity in vitro. In patients, significant improvements were observed in GVF from baseline ranging from 70% to 200% in three of five subjects aged 67-68 years, with largest improvements at 7-10 months. Of two GVF non-responders, one had significant visual acuity improvement (5-15 letters) from baseline after 6 months. CONCLUSION: Families with D477G variant have been identified in Ireland, the UK, France, the USA and Canada. Effects of single 7-day oral retinoid supplementation lasted at least 6 months, possibly giving visual benefit throughout remaining life in patients with advanced disease, where gene therapy is unlikely to prove beneficial.

11.
Life Sci Alliance ; 3(5)2020 05.
Article En | MEDLINE | ID: mdl-32312889

Retinal degeneration is the leading cause of incurable blindness worldwide and is characterised by progressive loss of light-sensing photoreceptors in the neural retina. SARM1 is known for its role in axonal degeneration, but a role for SARM1 in photoreceptor cell degeneration has not been reported. SARM1 is known to mediate neuronal cell degeneration through depletion of essential metabolite NAD and induction of energy crisis. Here, we demonstrate that SARM1 is expressed in photoreceptors, and using retinal tissue explant, we confirm that activation of SARM1 causes destruction of NAD pools in the photoreceptor layer. Through generation of rho -/- sarm1 -/- double knockout mice, we demonstrate that genetic deletion of SARM1 promotes both rod and cone photoreceptor cell survival in the rhodopsin knockout (rho -/- ) mouse model of photoreceptor degeneration. Finally, we demonstrate that SARM1 deficiency preserves cone visual function in the surviving photoreceptors when assayed by electroretinography. Overall, our data indicate that endogenous SARM1 has the capacity to consume NAD in photoreceptor cells and identifies a previously unappreciated role for SARM1-dependent cell death in photoreceptor cell degeneration.


Armadillo Domain Proteins/metabolism , Cytoskeletal Proteins/metabolism , Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , Animals , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/physiology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , Disease Models, Animal , Electroretinography , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NAD/metabolism , Photoreceptor Cells/physiology , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/metabolism , Rhodopsin/metabolism , Vision, Ocular
12.
JCI Insight ; 4(15)2019 08 08.
Article En | MEDLINE | ID: mdl-31391341

Age-related macular degeneration (AMD) is the leading cause of central retinal vision loss worldwide, with an estimated 1 in 10 people over the age of 55 showing early signs of the condition. There are currently no forms of therapy available for the end stage of dry AMD, geographic atrophy (GA). Here, we show that the inner blood-retina barrier (iBRB) is highly dynamic and may play a contributory role in GA development. We have discovered that the gene CLDN5, which encodes claudin-5, a tight junction protein abundantly expressed at the iBRB, is regulated by BMAL1 and the circadian clock. Persistent suppression of claudin-5 expression in mice exposed to a cholesterol-enriched diet induced striking retinal pigment epithelium (RPE) cell atrophy, and persistent targeted suppression of claudin-5 in the macular region of nonhuman primates induced RPE cell atrophy. Moreover, fundus fluorescein angiography in human and nonhuman primate subjects showed increased retinal vascular permeability in the evening compared with the morning. These findings implicate an inner retina-derived component in the early pathophysiological changes observed in AMD, and we suggest that restoring the integrity of the iBRB may represent a novel therapeutic target for the prevention and treatment of GA secondary to dry AMD.


ARNTL Transcription Factors/metabolism , Blood-Retinal Barrier/pathology , Circadian Clocks/physiology , Claudin-5/metabolism , Geographic Atrophy/pathology , Animals , Blood-Retinal Barrier/diagnostic imaging , Blood-Retinal Barrier/drug effects , Capillary Permeability/drug effects , Capillary Permeability/physiology , Chlorocebus aethiops , Claudin-5/genetics , Diet, High-Fat/adverse effects , Disease Models, Animal , Fluorescein Angiography , Fundus Oculi , Gene Knockdown Techniques , Geographic Atrophy/drug therapy , Geographic Atrophy/etiology , Geographic Atrophy/prevention & control , Healthy Volunteers , Humans , Magnetic Resonance Imaging , Mice , Mice, Transgenic , Photoperiod , RNA, Small Interfering/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology
13.
Prog Retin Eye Res ; 62: 120-133, 2018 01.
Article En | MEDLINE | ID: mdl-28951125

Protein levels of endothelial tight-junctions of the inner retinal microvasculature, together with those of Schlemm's canal, can be readily manipulated by RNA interference (RNAi), resulting in the paracellular clefts between such cells to be reversibly modulated. This facilitates access to the retina of systemically-deliverable low molecular weight, potentially therapeutic compounds, while also allowing potentially toxic material, for example, soluble Amyloid-ß1-40, to be removed from the retina into the peripheral circulation. The technique has also been shown to be highly effective in alleviation of pathological cerebral oedema and we speculate that it may therefore have similar utility in the oedematous retina. Additionally, by manipulating endothelial tight-junctions of Schlemm's canal, inflow of aqueous humour from the trabecular meshwork into the Canal can be radically enhanced, suggesting a novel avenue for control of intraocular pressure. Here, we review the technology underlying this approach together with specific examples of clinical targets that are, or could be, amenable to this novel form of genetic intervention.


Endothelium/physiology , Ocular Hypertension/physiopathology , Retinal Diseases/physiopathology , Tight Junctions/physiology , Trabecular Meshwork/physiology , Aqueous Humor/metabolism , Blood-Retinal Barrier/physiology , Humans , Retinal Vessels/physiology
14.
Expert Opin Ther Targets ; 21(11): 1037-1050, 2017 11.
Article En | MEDLINE | ID: mdl-28952395

INTRODUCTION: Ocular hypertension in open-angle glaucoma is caused by a reduced rate of removal of aqueous humour (AH) from the eye, with the majority of AH draining from the anterior chamber through the conventional outflow pathway, comprising the trabecular meshwork (TM) and Schlemm's Canal. Resistance to outflow is generated, in part, by the extracellular matrix (ECM) of the outflow tissues. Current pressure-lowering topical medications largely suppress AH production, or enhance its clearance through the unconventional pathway. However, therapies targeting the ECM of the conventional pathway in order to decrease intraocular pressure have become a recent focus of attention. Areas covered: We discuss the role of ECM of the TM in outflow homeostasis and its relevance as a target for glaucoma therapy, including progress in development of topical eye formulations, together with gene therapy approaches based on inducible, virally-mediated expression of matrix metalloproteinases to enhance aqueous outflow. Expert opinion: There remains a need for improved glaucoma medications that more specifically act upon sites causative to glaucoma pathogenesis. Emerging strategies targeting the ECM of the conventional outflow pathway, or associated components of the cytoskeleton of TM cells, involving new pharmacological formulations or genetically-based therapies, are promising avenues of future glaucoma treatment.


Drug Design , Genetic Therapy/methods , Glaucoma, Open-Angle/therapy , Molecular Targeted Therapy , Animals , Aqueous Humor/metabolism , Extracellular Matrix/metabolism , Glaucoma, Open-Angle/physiopathology , Humans , Intraocular Pressure , Matrix Metalloproteinases/metabolism , Trabecular Meshwork/metabolism
15.
Hum Mol Genet ; 26(R1): R2-R11, 2017 08 01.
Article En | MEDLINE | ID: mdl-28510639

While individually classed as rare diseases, hereditary retinal degenerations (IRDs) are the major cause of registered visual handicap in the developed world. Given their hereditary nature, some degree of intergenic heterogeneity was expected, with genes segregating in autosomal dominant, recessive, X-linked recessive, and more rarely in digenic or mitochondrial modes. Today, it is recognized that IRDs, as a group, represent one of the most genetically diverse of hereditary conditions - at least 260 genes having been implicated, with 70 genes identified in the most common IRD, retinitis pigmentosa (RP). However, targeted sequencing studies of exons from known IRD genes have resulted in the identification of candidate mutations in only approximately 60% of IRD cases. Given recent advances in the development of gene-based medicines, characterization of IRD patient cohorts for known IRD genes and elucidation of the molecular pathologies of disease in those remaining unresolved cases has become an endeavor of the highest priority. Here, we provide an outline of progress in this area.


Retinal Degeneration/genetics , Conserved Sequence , Exons , Eye Proteins/genetics , Humans , Mutation , Pedigree , Retinal Dystrophies/genetics , Retinitis Pigmentosa/genetics , Sequence Analysis, DNA
16.
Sci Rep ; 7: 40717, 2017 01 16.
Article En | MEDLINE | ID: mdl-28091584

The juxtacanalicular connective tissue of the trabecular meshwork together with inner wall endothelium of Schlemm's canal (SC) provide the bulk of resistance to aqueous outflow from the anterior chamber. Endothelial cells lining SC elaborate tight junctions (TJs), down-regulation of which may widen paracellular spaces between cells, allowing greater fluid outflow. We observed significant increase in paracellular permeability following siRNA-mediated suppression of TJ transcripts, claudin-11, zonula-occludens-1 (ZO-1) and tricellulin in human SC endothelial monolayers. In mice claudin-11 was not detected, but intracameral injection of siRNAs targeting ZO-1 and tricellulin increased outflow facility significantly. Structural qualitative and quantitative analysis of SC inner wall by transmission electron microscopy revealed significantly more open clefts between endothelial cells treated with targeting, as opposed to non-targeting siRNA. These data substantiate the concept that the continuity of SC endothelium is an important determinant of outflow resistance, and suggest that SC endothelial TJs represent a specific target for enhancement of aqueous movement through the conventional outflow system.


Anterior Chamber/physiology , Aqueous Humor/metabolism , Endothelium/metabolism , Tight Junctions/metabolism , Animals , Biomarkers , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Endothelium/ultrastructure , Gene Expression , Humans , Immunohistochemistry , Mice , Permeability , Primates , RNA Interference , RNA, Small Interfering/genetics , Tight Junctions/ultrastructure
17.
Br J Ophthalmol ; 100(4): 495-500, 2016 Apr.
Article En | MEDLINE | ID: mdl-26472407

BACKGROUND: The GNAT1 gene encodes the α subunit of the rod transducin protein, a key element in the rod phototransduction cascade. Variants in GNAT1 have been implicated in stationary night-blindness in the past, but unlike other proteins in the same pathway, it has not previously been implicated in retinitis pigmentosa. METHODS: A panel of 182 retinopathy-associated genes was sequenced to locate disease-causing mutations in patients with inherited retinopathies. RESULTS: Sequencing revealed a novel homozygous truncating mutation in the GNAT1 gene in a patient with significant pigmentary disturbance and constriction of visual fields, a presentation consistent with retinitis pigmentosa. This is the first report of a patient homozygous for a complete loss-of-function GNAT1 mutation. The clinical data from this patient provide definitive evidence of retinitis pigmentosa with late onset in addition to the lifelong night-blindness that would be expected from a lack of transducin function. CONCLUSION: These data suggest that some truncating GNAT1 variants can indeed cause a recessive, mild, late-onset retinal degeneration in human beings rather than just stationary night-blindness as reported previously, with notable similarities to the phenotype of the Gnat1 knockout mouse.


Codon, Nonsense , Heterotrimeric GTP-Binding Proteins/genetics , Retinitis Pigmentosa/genetics , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Base Sequence , DNA/isolation & purification , Electroretinography , Eye Diseases, Hereditary/diagnosis , Eye Diseases, Hereditary/genetics , Eye Diseases, Hereditary/physiopathology , Eye Proteins/genetics , Female , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/physiopathology , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Molecular Sequence Data , Myopia/diagnosis , Myopia/genetics , Myopia/physiopathology , Night Blindness/diagnosis , Night Blindness/genetics , Night Blindness/physiopathology , Retina/physiopathology , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/physiopathology , Siblings , Tomography, Optical Coherence , Transducin
20.
Hum Mol Genet ; 19(22): 4421-36, 2010 Nov 15.
Article En | MEDLINE | ID: mdl-20817636

Retinitis pigmentosa (RP) is the most prevalent cause of registered visual handicap among working aged populations of developed countries. Up to 40% of autosomal dominant cases of disease are caused by mutations within the rhodopsin, RDS-peripherin and inosine 5'-monophosphate dehydrogenase type 1 (IMPDH1) genes, at least 30 mutations within which give rise to proteins that cause disease pathology by misfolding and aggregation. Given the genetic complexity of this disease, therapies that simultaneously target multiple mutations are of substantial logistic and economic significance. We show here, in a murine model of autosomal dominant RP (RP10) involving expression of an Arg224Pro mutation within the IMPDH1 gene, that treatment with the low-molecular-weight drug, 17-allylamino-17-demethoxygeldanamycin (17-AAG), an ansamycin antibiotic that binds to heat shock protein Hsp90, activating a heat shock response in mammalian cells, protects photoreceptors against degeneration induced by aggregating mutant IMPDH1 protein, systemic delivery of this low-molecular-weight drug to the retina being facilitated by RNA interference-mediated modulation of the inner-blood retina barrier. 17-AAG has an orphan drug status and is in current clinical use for the treatment of non-ocular diseases. These data show that a single low-molecular-weight drug has the potential to suppress a wide range of mutant proteins causing RP.


Benzoquinones/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/therapeutic use , Retinitis Pigmentosa/prevention & control , Animals , Drug Delivery Systems , Drug Evaluation, Preclinical , Genes, Dominant , HSP90 Heat-Shock Proteins/genetics , HeLa Cells , Humans , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Mice , Mice, Inbred C57BL , Mutation , RNA Interference , Retina/drug effects , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Rhodopsin/genetics , Rhodopsin/metabolism
...