Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Mol Ther Methods Clin Dev ; 20: 86-94, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33376757

ABSTRACT

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.

2.
Genes (Basel) ; 11(12)2020 11 27.
Article in English | MEDLINE | ID: mdl-33261050

ABSTRACT

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.


Subject(s)
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
3.
BMJ Open Ophthalmol ; 5(1): e000462, 2020.
Article in English | MEDLINE | ID: mdl-32426524

ABSTRACT

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.

4.
Hum Mol Genet ; 26(R1): R2-R11, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28510639

ABSTRACT

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.


Subject(s)
Retinal Degeneration/genetics , Conserved Sequence , Exons , Eye Proteins/genetics , Humans , Mutation , Pedigree , Retinal Dystrophies/genetics , Retinitis Pigmentosa/genetics , Sequence Analysis, DNA
5.
Hum Mol Genet ; 26(7): 1230-1246, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28158775

ABSTRACT

Intraocular pressure (IOP) is maintained as a result of the balance between production of aqueous humour (AH) by the ciliary processes and hydrodynamic resistance to its outflow through the conventional outflow pathway comprising the trabecular meshwork (TM) and Schlemm's canal (SC). Elevated IOP, which can be caused by increased resistance to AH outflow, is a major risk factor for open-angle glaucoma. Matrix metalloproteinases (MMPs) contribute to conventional aqueous outflow homeostasis in their capacity to remodel extracellular matrices, which has a direct impact on aqueous outflow resistance and IOP. We observed decreased MMP-3 activity in human glaucomatous AH compared to age-matched normotensive control AH. Treatment with glaucomatous AH resulted in significantly increased transendothelial resistance of SC endothelial and TM cell monolayers and reduced monolayer permeability when compared to control AH, or supplemented treatment with exogenous MMP-3.Intracameral inoculation of AAV-2/9 containing a CMV-driven MMP-3 gene (AAV-MMP-3) into wild type mice resulted in efficient transduction of corneal endothelium and an increase in aqueous concentration and activity of MMP-3. Most importantly, AAV-mediated expression of MMP-3 increased outflow facility and decreased IOP, and controlled expression using an inducible promoter activated by topical administration of doxycycline achieved the same effect. Ultrastructural analysis of MMP-3 treated matrices by transmission electron microscopy revealed remodelling and degradation of core extracellular matrix components. These results indicate that periodic induction, via use of an eye drop, of AAV-mediated secretion of MMP-3 into AH could have therapeutic potential for those cases of glaucoma that are sub-optimally responsive to conventional pressure-reducing medications.


Subject(s)
Dependovirus/genetics , Glaucoma/therapy , Intraocular Pressure/genetics , Matrix Metalloproteinase 3/genetics , Animals , Aqueous Humor/metabolism , Disease Models, Animal , Endothelium, Corneal/metabolism , Endothelium, Corneal/pathology , Glaucoma/genetics , Glaucoma/pathology , Humans , Matrix Metalloproteinase 3/therapeutic use , Mice , Ophthalmic Solutions/therapeutic use
6.
Sci Rep ; 7: 40717, 2017 01 16.
Article in English | MEDLINE | ID: mdl-28091584

ABSTRACT

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.


Subject(s)
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
7.
Sci Transl Med ; 6(230): 230ra44, 2014 Apr 02.
Article in English | MEDLINE | ID: mdl-24695684

ABSTRACT

Age-related macular degeneration (AMD) is the most common form of central retinal blindness globally. Distinct processes of the innate immune system, specifically activation of the NLRP3 inflammasome, have been shown to play a central role in the development of both "dry" and neovascular ("wet") forms of the disease. We show that the inflammatory cytokine interleukin-18 (IL-18) can regulate choroidal neovascularization formation in mice. We observed that exogenous administration of mature recombinant IL-18 has no effect on retinal pigment epithelial (RPE) cell viability, but that overexpression of pro-IL-18 or pro-IL-1ß alone can cause RPE cell swelling and subsequent atrophy, a process that can be inhibited by the promotion of autophagy. A direct comparison of local and systemic administration of mature recombinant IL-18 with current anti-VEGF (vascular endothelial growth factor)-based therapeutic strategies shows that IL-18 treatment works effectively alone and more effectively in combination with anti-VEGF therapy and represents a novel therapeutic strategy for the treatment of wet AMD.


Subject(s)
Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/prevention & control , Interleukin-18/therapeutic use , Macular Degeneration/drug therapy , Animals , Autophagy/drug effects , Cell Line , Cell Survival/drug effects , Choroidal Neovascularization/complications , Choroidal Neovascularization/pathology , Hematopoiesis/drug effects , Humans , Interleukin-18/pharmacology , Interleukin-1beta/pharmacology , Interleukin-1beta/therapeutic use , Intravitreal Injections , Lasers , Macular Degeneration/complications , Macular Degeneration/pathology , Mice , Models, Biological , Permeability/drug effects , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
8.
Adv Exp Med Biol ; 801: 229-35, 2014.
Article in English | MEDLINE | ID: mdl-24664703

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of legal blindness in elderly individuals in the developed world, affecting 30-50 million people worldwide. AMD primarily affects the macular region of the retina that is responsible for the majority of central, color and daytime vision. The presence of drusen, extracellular protein aggregates that accumulate under the retinal pigment epithelium (RPE), is a major pathological hallmark in the early stages of the disease. The end stage 'dry' and 'wet' forms of the disease culminate in vision loss and are characterized by focal degeneration of the RPE and cone photoreceptors, and choroidal neovascularization (CNV), respectively. Being a multifactorial and genetically heterogeneous disease, the pathophysiology of AMD remains unclear, yet, there is ample evidence supporting immunological and inflammatory processes. Here, we review the recent literature implicating some of these immune processes in human AMD and in animal models.


Subject(s)
Inflammation/immunology , Macular Degeneration/immunology , Optic Disk Drusen/immunology , Retinitis/immunology , Signal Transduction/immunology , Humans
9.
Adv Exp Med Biol ; 801: 409-15, 2014.
Article in English | MEDLINE | ID: mdl-24664725

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of central vision loss worldwide and while polymorphisms in genes associated with the immune system have been identified as risk factors for disease development, the underlying pathways and mechanisms involved in disease progression have remained unclear. In AMD, localised inflammatory responses related to particulate matter accumulation and subsequent "sterile" inflammation has recently gained considerable interest amongst basic researchers and clinicians alike. Typically, inflammatory responses in the human body are caused as a result of bacterial or viral infection, however in chronic conditions such as AMD, extracellular particulate matter such as drusen can be "sensed" by the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome, culminating in the release of the two pro-inflammatory cytokines IL-1ß and IL-18 in the delicate local tissue of the retina. Identification at the molecular level of mediators of the inflammatory response in AMD may yield novel therapeutic approaches to this common and often severe form of blindness. Here, we will describe the role of IL-18 in AMD and other forms of retinal disorders. We will outline some of the key functions of IL-18 as it pertains to maintaining tissue homeostasis in a healthy and degenerating/diseased retina.


Subject(s)
Inflammasomes/immunology , Interleukin-18/immunology , Macular Degeneration/immunology , Retinal Degeneration/immunology , Retinitis/immunology , Humans
10.
Adv Exp Med Biol ; 801: 471-6, 2014.
Article in English | MEDLINE | ID: mdl-24664733

ABSTRACT

The first autosomal dominant mutation identified to cause retinitis pigmentosa in the North American population was the substitution of proline to histidine at position 23 of the rhodopsin gene (P23H RHO). Many biochemical studies have demonstrated that P23H mutation induces rhodopsin (RHO) misfolding leading to endoplasmic reticulum stress. Herein, we review current thinking of this topic.


Subject(s)
Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/therapy , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Rhodopsin/genetics , Animals , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Genes, Dominant , Humans
11.
Adv Exp Med Biol ; 801: 783-9, 2014.
Article in English | MEDLINE | ID: mdl-24664771

ABSTRACT

Disease mechanisms associated with retinal disease are of immense complexity, mutations within 45 genes having been implicated, for example, in retinitis pigmentosa, while interplay between genetic, environmental, and demographic factors can lead to diabetic retinopathy, age-related macular degeneration, and glaucoma. In light of such diversity, any therapeutic modality that can be targeted to an early molecular process instrumental in multiple forms of disease, such as oxidative stress, holds much attraction. Here, we provide a brief overview of a selection of compounds displaying antioxidant activity, which have been shown to slow down degeneration of retinal tissues and highlight suggested modes of action.


Subject(s)
Antioxidants/pharmacology , Oxidative Stress/drug effects , Plant Proteins/pharmacology , Retinal Diseases/drug therapy , Animals , Humans
12.
Nat Commun ; 3: 849, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22617289

ABSTRACT

Traumatic brain injury is the leading cause of death in children and young adults globally. Malignant cerebral oedema has a major role in the pathophysiology that evolves after severe traumatic brain injury. Added to this is the significant morbidity and mortality from cerebral oedema associated with acute stroke, hypoxic ischemic coma, neurological cancers and brain infection. Therapeutic strategies to prevent cerebral oedema are limited and, if brain swelling persists, the risks of permanent brain damage or mortality are greatly exacerbated. Here we show that a temporary and size-selective modulation of the blood-brain barrier allows enhanced movement of water from the brain to the blood and significantly impacts on brain swelling. We also show cognitive improvement in mice with focal cerebral oedema following administration in these animals of short interfering RNA directed against claudin-5. These observations may have profound consequences for early intervention in cases of traumatic brain injury, or indeed any neurological condition where cerebral oedema is the hallmark pathology.


Subject(s)
Brain Edema/etiology , Brain Edema/therapy , Brain Injuries/complications , Claudins/metabolism , Cognition/physiology , Animals , Blood-Brain Barrier/metabolism , Brain Edema/diagnostic imaging , Brain Injuries/diagnostic imaging , Child , Claudin-5 , Claudins/genetics , Humans , Intracranial Pressure/physiology , Male , Mice , RNA Interference , Tomography, X-Ray Computed
13.
Nat Med ; 18(5): 791-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22484808

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of central vision loss worldwide. Drusen accumulation is the major pathological hallmark common to both dry and wet AMD. Although activation of the immune system has been implicated in disease progression, the pathways involved are unclear. Here we show that drusen isolated from donor AMD eyes activates the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome, causing secretion of interleukin-1b (IL-1b) and IL-18. Drusen component C1Q also activates the NLRP3 inflammasome. Moreover, the oxidative-stress-related protein-modification carboxyethylpyrrole (CEP), a biomarker of AMD, primes the inflammasome. We found cleaved caspase-1 and NLRP3 in activated macrophages in the retinas of mice immunized with CEP-adducted mouse serum albumin, modeling a dry-AMD­like pathology. We show that laser-induced choroidal neovascularization (CNV), a mouse model of wet AMD, is exacerbated in Nlrp3(-/-) but not Il1r1(-/-) mice, directly implicating IL-18 in the regulation of CNV development. These findings indicate a protective role for NLRP3 and IL-18 in the progression of AMD.


Subject(s)
Carrier Proteins/physiology , Interleukin-18/physiology , Macular Degeneration/prevention & control , Optic Disk Drusen/metabolism , Animals , Cells, Cultured , Choroidal Neovascularization/etiology , Choroidal Neovascularization/prevention & control , Complement C1q/physiology , Immunization , Interleukin-1beta/physiology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Phagosomes/physiology
14.
Eur J Hum Genet ; 20(1): 64-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21863053

ABSTRACT

Retinitis pigmentosa (RP) is a degenerative retinal disease involving progressive loss of rod and cone photoreceptor function. It represents the most common form of registered blindness among the working aged populations of developed countries. Given the immense genetic heterogeneity associated with this disease, parameters influencing cone photoreceptor survival (preservation of daytime vision) that are independent of primary mutations are exceedingly important to identify from a therapeutic standpoint. Here we identify C1q, the primary component of the classical complement pathway, as a cone photoreceptor neuronal survival factor.


Subject(s)
Complement C1q/metabolism , Retina/pathology , Retinal Cone Photoreceptor Cells/pathology , Retinitis Pigmentosa/pathology , Animals , Cell Survival , Complement Pathway, Classical , Disease Models, Animal , Electroretinography , Genotype , Immunohistochemistry , Membrane Glycoproteins/metabolism , Mice , Mice, Congenic , Mice, Inbred C57BL , Receptors, Complement/metabolism , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinitis Pigmentosa/metabolism
18.
EMBO Mol Med ; 3(4): 235-45, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21374818

ABSTRACT

We describe a procedure for controlled, periodic, reversible modulation of selected regions of the blood-brain barrier (BBB) or the inner-blood-retina barrier (iBRB) based on incorporation into an AAV-2/9 vector of a doxycycline-inducible gene encoding shRNA targeting claudin-5, one of 30 or so proteins constituting the BBB and iBRB. The vector may be introduced stereotaxically into pre-selected regions of the brain or into the retina, rendering these regions permeable to low-molecular weight compounds up to approximately 1 kDa for the period of time during which the inducing agent, doxycycline, is administered in drinking water, but excluding potentially toxic higher molecular weight materials. We report on the use of barrier modulation in tandem with systemic drug therapy to prevent retinal degeneration and to suppress laser-induced choroidal neovascularization (CNV), the latter being the hallmark pathology associated with the exudative, or wet, form of age-related macular degeneration (AMD). These observations constitute the basis of a minimally invasive systemic therapeutic modality for retinal diseases, including retinitis pigmentosa and AMD, where, in early stage disease, the iBRB is intact and impervious to systemically administered drugs.


Subject(s)
Blood-Retinal Barrier/drug effects , Drug Delivery Systems/methods , Genetic Therapy , Macular Degeneration/therapy , Animals , Blood-Retinal Barrier/innervation , Blood-Retinal Barrier/metabolism , Cell Line , Claudin-5 , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Macular Degeneration/genetics , Macular Degeneration/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Weight , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use
19.
Hum Mol Genet ; 19(22): 4421-36, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20817636

ABSTRACT

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.


Subject(s)
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
20.
Adv Exp Med Biol ; 664: 301-8, 2010.
Article in English | MEDLINE | ID: mdl-20238029

ABSTRACT

The inner Blood-Retina-barrier (iBRB) remains a key element in retarding the development of novel therapeutics for the treatment of many ocular disorders. The iBRB contains tight-junctions (TJ's) which reduce the space between adjacent endothelial cells lining the fine capillaries of the retinal microvasculature to form a selective and regulatable barrier. We have recently shown that in mice, the iBRB can be transiently and size-selectively opened to molecules with molecular weights of up to approximately 1 kDa using an siRNA-mediated approach involving suppression of the tight junction protein, claudin-5. We have systemically delivered siRNA targeting claudin-5 to retinal capillary endothelial cells in mice and through a series of tracer experiments and magnetic-resonance-imaging (MRI), we have shown a transient and size-selective increase in permeability at the iBRB to molecules below 1 kDa. The potential to exploit this specific compromise in iBRB integrity may have far reaching implications for the development of experimental animal models of retinal degenerative disorders, and for enhanced delivery of therapeutic molecules which would normally not traverse the iBRB. Using RNAi-mediated opening of the iBRB, the systemic delivery of low molecular weight therapeutics could in principle, hold real promise as an alternative to repeated intraocular inoculation of compounds. Results demonstrated here in mouse models, should lead to a 'humanized' form of systemic delivery as opposed to the hydrodynamic approach used in our work to date.


Subject(s)
Blood-Retinal Barrier/metabolism , Eye Diseases/therapy , Animals , Benzimidazoles/metabolism , Claudin-5 , Cryoultramicrotomy , Extravasation of Diagnostic and Therapeutic Materials , Gadolinium DTPA/metabolism , Injections , Magnetic Resonance Imaging , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Molecular Weight , Perfusion , RNA Interference , RNA, Small Interfering/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...