Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Phlebology ; : 2683555241239721, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39175283

ABSTRACT

Dermatophlebology refers to the study of dermal alterations that occur with transmitted high venous pressure. These changes may range from telangiectasia, complex dermal vascular conditions, and all changes in Clinical Etiology Anatomical Pathophysiology classes IV-VI. Understanding the underlying pathophysiology is essential to treating the skin manifestations of venous disease.

2.
Transl Vis Sci Technol ; 9(7): 28, 2020 06.
Article in English | MEDLINE | ID: mdl-32844051

ABSTRACT

Purpose: Electric micro-current has been shown to enhance penetration and transduction of adeno-associated viral (AAV) vectors in mouse retina after intravitreal administration. We termed this: "electric-current vector mobility (ECVM)." The present study considered whether ECVM could augment retinal transduction efficiency of intravitreal AAV8-CMV-EGFP in normal rabbit and nonhuman primate (NHP) macaque. Potential mechanisms underlying enhanced retinal transduction by ECVM were also studied. Methods: We applied an electric micro-current across the intact eye of normal rabbit and monkey in vivo for a brief period immediately after intravitreal injection of AAV8-CMV-EGFP. Retinal GFP expression was evaluated by fundus imaging in vivo. Retinal immunohistochemistry was performed to assess the distribution of retinal cells transduced by the AAV8-EGFP. Basic fibroblast growth factor (bFGF) was analyzed by quantitative RT-polymerase chain reaction (PCR). Müller glial reactivity and inner limiting membrane (ILM) were examined by the glial fibrillary acidic protein (GFAP) and vimentin staining in mouse retina, respectively. Results: ECVM significantly increased the efficiency of AAV reaching and transducing the rabbit retina following intravitreal injection, with gene expression in inner nuclear layer, ganglion cells, and Müller cells. Similar trend of improvement was observed in the ECVM-treated monkey eye. The electric micro-current upregulated bFGF expression in Müller cells and vimentin showed ILM structural changes in mouse retina. Conclusions: ECVM promotes the transduction efficiency of AAV8-CMV-GFP in normal rabbit and monkey retinas following intravitreal injection. Translational Relevance: This work has potential translational relevance to human ocular gene therapy by increasing retinal expression of therapeutic vectors given by intravitreal administration.


Subject(s)
Dependovirus , Genetic Vectors , Animals , Dependovirus/genetics , Gene Expression , Genetic Vectors/genetics , Rabbits , Retina , Transduction, Genetic
3.
Invest Ophthalmol Vis Sci ; 60(10): 3680-3688, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31469895

ABSTRACT

Purpose: The b-wave of the cone ERG increases in amplitude and speed during the first few minutes of adaptation to a rod-suppressing background light. Earlier studies implicate rod pathway input to the cone pathway in these changes. Methods: The timing and amplitude of the cone b-wave and isolated oscillatory potentials (OP) during the first 10 minutes of light adaptation in wild-type (WT) mice and two mutant lines without functional rods was examined: rhodopsin knockout (Rho-/-), lacking rod outer segments, and NRL knockout (Nrl-/-), in which rods are replaced by S-cones. Expression of the immediate-early gene c-fos, which is increased in the inner retina by light-induced activity, was evaluated by immunohistochemistry in dark- and light-adapted retinas. Results: WT b-wave and OP amplitudes increased, and implicit times decreased during light adaptation. Subtracting OP did not alter b-wave changes. Rho-/- b-wave and OP amplitudes did not increase during adaptation. B-wave timing and amplitude and the timing of the major OP at 1 minute of adaptation were equivalent to WT at 10 minutes. The light-adapted ERG b-wave in Nrl-/- mice, which originates in both the rod and cone pathways, changed in absolute amplitude and timing similar to WT. C-fos expression was present in the inner retinas of dark-adapted Rho-/- but not WT or Nrl-/- mice. Conclusions: Activity in the distal rod pathway produces changes in the cone ERG during light adaptation. Rods in Rho-/- mice constitutively activate this rod-cone pathway interaction. The rod pathway S-cones in Nrl-/- mice may maintain the WT interaction.


Subject(s)
Adaptation, Ocular/physiology , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Electroretinography , Eye Proteins/genetics , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Photic Stimulation , Proto-Oncogene Proteins c-fos/genetics , Retina/physiology , Rhodopsin/genetics
4.
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
5.
Mol Ther Methods Clin Dev ; 13: 77-85, 2019 Jun 14.
Article in English | MEDLINE | ID: mdl-30719486

ABSTRACT

Adeno-associated virus (AAV) vector-mediated gene delivery is a promising approach for therapy, but implementation in the eye currently is hampered by the need for delivering the vector underneath the retina, using surgical application into the subretinal space. This limits the extent of the retina that is treated and may cause surgical injury. Vector delivery into the vitreous cavity would be preferable because it is surgically less invasive and would reach more of the retina. Unfortunately, most conventional, non-modified AAV vector serotypes penetrate the retina poorly from the vitreous; this limits efficient transduction and expression by target cells (retinal pigment epithelium and photoreceptors). We developed a method of applying a small and safe electric current across the intact eye in vivo for a brief period following intravitreal vector administration. This significantly improved AAV-mediated transduction of retinal cells in wild-type mice following intravitreal delivery, with gene expression in retinal pigment epithelium and photoreceptor cells. The low-level current had no adverse effects on retinal structure and function. This method should be generally applicable for other AAV serotypes and may have broad application in both basic research and clinical studies.

6.
Mol Ther ; 26(9): 2282-2294, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30196853

ABSTRACT

This study evaluated the safety and tolerability of ocular RS1 adeno-associated virus (AAV8-RS1) gene augmentation therapy to the retina of participants with X-linked retinoschisis (XLRS). XLRS is a monogenic trait affecting only males, caused by mutations in the RS1 gene. Retinoschisin protein is secreted principally in the outer retina, and its absence results in retinal cavities, synaptic dysfunction, reduced visual acuity, and susceptibility to retinal detachment. This phase I/IIa single-center, prospective, open-label, three-dose-escalation clinical trial administered vector to nine participants with pathogenic RS1 mutations. The eye of each participant with worse acuity (≤63 letters; Snellen 20/63) received the AAV8-RS1 gene vector by intravitreal injection. Three participants were assigned to each of three dosage groups: 1e9 vector genomes (vg)/eye, 1e10 vg/eye, and 1e11 vg/eye. The investigational product was generally well tolerated in all but one individual. Ocular events included dose-related inflammation that resolved with topical and oral corticosteroids. Systemic antibodies against AAV8 increased in a dose-related fashion, but no antibodies against RS1 were observed. Retinal cavities closed transiently in one participant. Additional doses and immunosuppressive regimens are being explored to pursue evidence of safety and efficacy (ClinicalTrials.gov: NCT02317887).


Subject(s)
Eye Proteins/metabolism , Genetic Therapy/methods , Retinoschisis/therapy , Adult , Aged , Eye Proteins/genetics , Female , Humans , Intravitreal Injections , Male , Middle Aged , Mutation/genetics , Retina/metabolism , Retina/pathology , Retinoschisis/genetics , Retinoschisis/metabolism , Young Adult
7.
J Cosmet Dermatol ; 17(5): 779-782, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30226025

ABSTRACT

BACKGROUND: Heat modalities are commonly used as either primary or adjunctive treatment for telangiectasia. Minimal information is available as to the nature of injury to the vessel and surrounding tissue. METHOD: A total of 135 patients were treated over a 2-year period using ohmic thermolysis (45), 940 nm laser (50), and 940 nm laser with sclerotherapy (40). After treatment, 1 mm biopsies were done in selected patients in each group. Clinical correlation was studied in each group by observing vessel response at 4-6 weeks postprocedure. RESULTS: Ohmic thermolysis produces electrodessication of the squamous epithelium, reticular dermis, and fusion of the target vessel. 940 nm laser results include squamous epithelial damage, subcutaneous water blister, collagen denaturation, and vessel endothelial cell loss with thrombus at point of maximal impact. The addition of sclerotherapy at time of laser potentiates vessel damage. There was no long-term skin sequelae after treatment when each device is used at recommended settings and on appropriate vessel size. CONCLUSION: Each device causes damage to the squamous epithelium and papillary reticular dermis that is transient. Ohmic thermolysis provides vessel clearance of >90% in telangiectasias <0.5 mm. 940 nm laser effectiveness is <70% for vessel clearance, but improves to >90% when sclerotherapy is performed at time of treatment.


Subject(s)
Electrocoagulation/methods , Laser Coagulation/methods , Radiofrequency Therapy/methods , Telangiectasis/pathology , Telangiectasis/therapy , Humans , Sclerotherapy , Skin/pathology
8.
10.
Invest Ophthalmol Vis Sci ; 58(3): 1656-1664, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28297725

ABSTRACT

Purpose: To test the effects of rearing light intensity on retinal function and morphology in the retinoschisis knockout (Rs1-KO) mouse model of X-linked retinoschisis, and whether it affects functional outcome of RS1 gene replacement. Methods: Seventy-six Rs1-KO mice were reared in either cyclic low light (LL, 20 lux) or moderate light (ML, 300 lux) and analyzed at 1 and 4 months. Retinal function was assessed by electroretinogram and cavity size by optical coherence tomography. Expression of inward-rectifier K+ channel (Kir4.1), water channel aquaporin-4 (AQP4), and glial fibrillary acidic protein (GFAP) were analyzed by Western blotting. In a separate study, Rs1-KO mice reared in LL (n = 29) or ML (n = 27) received a unilateral intravitreal injection of scAAV8-hRs-IRBP at 21 days, and functional outcome was evaluated at 4 months by electroretinogram. Results: At 1 month, no functional or structural differences were found between LL- or ML-reared Rs1-KO mice. At 4 months, ML-reared Rs1-KO mice showed significant reduction of b-wave amplitude and b-/a-wave ratio with no changes in a-wave, and a significant increase in cavity size, compared to LL-reared animals. Moderate light rearing increased Kir4.1 expression in Rs1-KO mice by 4 months, but not AQP4 and GFAP levels. Administration of scAAV8-hRS1-IRBP to Rs1-KO mice showed similar improvement of inner retinal ERG function independent of LL or ML rearing. Conclusions: Rearing light conditions affect the development of retinal cavities and post-photoreceptor function in Rs1-KO mice. However, the effect of rearing light intensity does not interact with the efficacy of RS1 gene replacement in Rs1-KO mice.


Subject(s)
Genetic Therapy/methods , Light , Retinal Photoreceptor Cell Inner Segment/pathology , Retinoschisis/therapy , Animals , Blotting, Western , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Electroretinography , Eye Proteins/genetics , Eye Proteins/metabolism , Follow-Up Studies , Gene Expression Regulation , Gene Transfer Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Potassium Channels, Inwardly Rectifying/biosynthesis , Potassium Channels, Inwardly Rectifying/genetics , RNA/genetics , Retinal Photoreceptor Cell Inner Segment/radiation effects , Retinoschisis/diagnosis , Retinoschisis/genetics , Retinoschisis/physiopathology , Time Factors , Tomography, Optical Coherence
11.
Phlebology ; 32(1_suppl): 3-19, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28211296

ABSTRACT

Scope A systematic review of the clinical literature concerning medical management of chronic venous disease with the venoactive therapy Micronized Purified Flavonoid Fraction was conducted in addition to an investigation of the hemodynamics and mechanism of chronic venous disease. Methods The systematic review of the literature focused on the use of Micronized Purified Flavonoid Fraction (diosmin) which has recently become available in the US, in the management of chronic venous disease. The primary goal was to assess the level of evidence of the role of Micronized Purified Flavonoid Fraction in the healing of ulcers, and secondarily on the improvement of the symptoms of chronic venous disease such as edema. An initial search of Medline, Cochrane Database for Systematic Reviews and Google Scholar databases was conducted. The references of articles obtained in the primary search, including a Cochrane review of phlebotonics for venous insufficiency, were reviewed for additional studies. Studies were included if patients had a diagnosis of chronic venous disease documented with Doppler and Impedance Plethysmography. Studies excluded were those that had patients with arterial insufficiency (Ankle Brachial Index < .6), comorbidity of diabetes, obesity, rheumatological diseases, or if other causes of edema were present (congestive heart failure, renal, hepatic or lymphatic cause), or if the patient population had recent surgery or deep vein thrombosis, or had been using diuretics (in studies of edema). Other elements of the study design were to note specifically the type of compression therapy used in conjunction with Micronized Purified Flavonoid Fraction. Results The literature review yielded 250 abstracts, 65 of which met criteria for further review and 10 papers were selected for consideration in the systematic review. Conclusion In summary, the general level of evidence supports the recommendation that the use of medical therapy with Micronized Purified Flavonoid Fraction has beneficial outcomes without serious adverse events. In the United States, diosmiplex is the only available prescription formulation of Micronized Purified Flavonoid Fraction. It is derived from the rinds of oranges and is categorized as a medical food and not as a drug; and may be a particularly attractive therapy for many chronic venous disease patients because of its favorable safety profile. The Working Group for chronic venous disease concurs with previous guidance by the International European Society for Vascular Surgery in 2015 which recommended the use of Micronized Purified Flavonoid Fraction for the healing of venous ulcers, alone and adjunctive to compression therapy, and for the reduction in symptoms of chronic venous disease such as edema.


Subject(s)
Flavonoids/therapeutic use , Vascular Diseases/drug therapy , Vascular Diseases/epidemiology , Animals , Capillaries/cytology , Cardiology/methods , Chronic Disease , Edema/therapy , Hemodynamics , Humans , Hypertension , Leukocytes/cytology , Permeability , Quality of Life , Varicose Ulcer/epidemiology , Varicose Ulcer/physiopathology , Vascular Diseases/physiopathology , Veins/physiopathology , Venous Insufficiency/epidemiology , Venous Insufficiency/physiopathology , Venous Thrombosis/drug therapy , Venous Thrombosis/epidemiology
12.
Phlebology ; 32(7): 496-500, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27738241

ABSTRACT

Introduction This study was designed to determine by histological evaluation and clinical correlation the most effective sclerosant concentration of Sotradecol® (sodium tetradecyl sulfate) and Asclera® (polidocanol) for the treatment of leg telangiectasia. Methods Histological studies were completed on 40 patients, all of whom were female with a mean age of 53. After sclerotherapy with varying concentrations of sclerosant solutions for the treatment of 0.8 mm and 1 mm leg telangiectasia, histological specimens were examined for the following criteria: luminal changes, subintimal changes, smooth muscle wall alterations, and vessel wall integrity. Six patients from this group were also treated with sodium tetradecyl sulfate foam 0.1%-0.2% or polidocanol foam 0.31% for the treatment of 2 mm reticular veins. In a second group of 20 patients, clinical results after treatment with sodium tetradecyl sulfate 0.15% and polidocanol 0.31% were evaluated. Results Histological findings in patients treated with sodium tetradecyl sulfate 0.05% and polidocanol 0.25% were minimal. Sclerosant concentrations of sodium tetradecyl sulfate ≥0.2% revealed intraluminal debris and greater smooth wall damage. Polidocanol 0.5% was equivalent in strength to sodium tetradecyl sulfate 0.2% on histological evaluation. Sodium tetradecyl sulfate 0.15% and polidocanol 0.31% had similar findings on histological examination with mild smooth muscle wall changes, endothelial cell lysis, and subintimal layer damage. Reticular veins treated with 0.1%, 0.15%, and 0.2% sodium tetradecyl sulfate foam and 0.31% polidocanol foam produced identical histological findings. Conclusion Sodium tetradecyl sulfate 0.15% and polidocanol 0.31% based on histological evaluations and clinical correlation are the best sclerosant concentrations for 0.8 mm to 1 mm leg telangiectasia. Sodium tetradecyl sulfate foam is comparable to polidocanol foam at these concentrations as well.


Subject(s)
Leg/pathology , Polyethylene Glycols/administration & dosage , Sclerosing Solutions/administration & dosage , Sclerotherapy , Sodium Tetradecyl Sulfate/administration & dosage , Telangiectasis/therapy , Cohort Studies , Endothelial Cells , Female , Humans , Hyperpigmentation , Inflammation , Middle Aged , Muscle, Smooth/pathology , Polidocanol , Veins/pathology
13.
Mol Ther Methods Clin Dev ; 5: 16011, 2016.
Article in English | MEDLINE | ID: mdl-27626041

ABSTRACT

X-linked retinoschisis (XLRS) is a retinal disease caused by mutations in the gene encoding the protein retinoschisin (RS1) and is one of the most common causes of macular degeneration in young men. Our therapeutic approach for XLRS is based on the administration of AAV8-scRS/IRBPhRS, an adeno-associated viral vector coding the human RS1 protein, via the intravitreal (IVT) route. Two Good Laboratory Practice studies, a 9-month study in New Zealand White rabbits (n = 124) injected with AAV8-scRS/IRBPhRS at doses of 2E9, 2E10, 2E11, and 1.5E12 vector genomes/eye (vg/eye), and a 6-month study in Rs1-KO mice (n = 162) dosed with 2E9 and 2E10 vg/eye of the same vector were conducted to assess ocular and systemic safety. A self-resolving, dose-dependent vitreal inflammation was the main ocular finding, and except for a single rabbit dosed with 1.5E12 vg/eye, which showed a retinal detachment, no other ocular adverse event was reported. Systemic toxicity was not identified in either species. Biodistribution analysis in Rs1-KO mice detected spread of vector genome in extraocular tissues, but no evidence of organ or tissues damage was found. These studies indicate that IVT administration of AAV8-scRS/IRBPhRS is safe and well tolerated and support its advancement into a phase 1/2a clinical trial for XLRS.

14.
Invest Ophthalmol Vis Sci ; 57(9): OCT277-87, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27409484

ABSTRACT

PURPOSE: Spectral-domain optical coherence tomography (SD-OCT) was used to characterize the retinal phenotype, natural history, and treatment responses in a mouse model of X-linked retinoschisis (Rs1-KO) and to identify new structural markers of AAV8-mediated gene therapy outcome. METHODS: Optical coherence tomography scans were performed on wild-type and Rs1-KO mouse retinas between 1 and 12 months of age and on Rs1-KO mice after intravitreal injection of AAV8-scRS/IRBPhRS (AAV8-RS1). Cavities and photoreceptor outer nuclear layer (ONL) thickness were measured, and outer retina reflective band (ORRB) morphology was examined with age and after AAV8-RS1 treatment. Outer retina reflective band morphology was compared to immunohistochemical staining of the outer limiting membrane (OLM) and photoreceptor inner segment (IS) mitochondria and to electron microscopy (EM) images of IS. RESULTS: Retinal cavity size in Rs1-KO mice increased between 1 and 4 months and decreased thereafter, while ONL thickness declined steadily, comparable to previous histologic studies. Wild-type retina had four ORRBs. In Rs1-KO, ORRB1was fragmented from 1 month, but was normal after 8 months; ORRB2 and ORRB3 were merged at all ages. Outer retina reflective band morphology returned to normal after AAV-RS1 therapy, paralleling the recovery of the OLM and IS mitochondria as indicated by anti-ß-catenin and anti-COX4 labeling, respectively, and EM. CONCLUSIONS: Spectral-domain OCT is a sensitive, noninvasive tool to monitor subtle changes in retinal morphology, disease progression, and effects of therapies in mouse models. The ORRBs may be useful to assess the outcome of gene therapy in the treatment of X-linked retinoschisis patients.


Subject(s)
Cell Adhesion Molecules/deficiency , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Retina/ultrastructure , Retinoschisis/diagnosis , Tomography, Optical Coherence/methods , Animals , Disease Models, Animal , Electroretinography , Eye Proteins , Intravitreal Injections , Mice , Mice, Knockout , Microscopy, Immunoelectron , Retinoschisis/genetics , Retinoschisis/metabolism
15.
Invest Ophthalmol Vis Sci ; 57(6): 2864-75, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27233035

ABSTRACT

PURPOSE: The active form of small GTPase RAC1 is required for activation of NADPH oxidase (NOX), which in turn generates reactive oxygen species (ROS) in nonphagocytic cells. We explored whether NOX-induced oxidative stress contributes to rod degeneration in retinas expressing constitutively active (CA) RAC1. METHODS: Transgenic (Tg)-CA-RAC1 mice were given apocynin (10 mg/kg, intraperitoneal), a NOX inhibitor, or vehicle daily for up to 13 weeks. Superoxide production and oxidative damage were assessed by dihydroethidium staining and by protein carbonyls and malondialdehyde levels, respectively. Outer nuclear layer (ONL) cells were counted and electroretinogram (ERG) amplitudes measured in Tg-CA-RAC1 mice. Outer nuclear layer cells were counted in wild-type (WT) mice after transfer of CA-Rac1 gene by subretinal injection of AAV8-pOpsin-CA Rac1-GFP. RESULTS: Transgenic-CA-RAC1 retinas had significantly fewer photoreceptor cells and more apoptotic ONL cells than WT controls from postnatal week (Pw) 3 to Pw13. Superoxide accumulation and protein and lipid oxidation were increased in Tg-CA-RAC1 retinas and were reduced in mice treated with apocynin. Apocynin reduced the loss of photoreceptors and increased the rod ERG a- and b-wave amplitudes when compared with vehicle-injected transgenic controls. Photoreceptor loss was also observed in regions of adult WT retina transduced with AAV8-pOpsin-CA Rac1-GFP but not in neighboring regions that were not transduced or in AAV8-pOpsin-GFP-transduced retinas. CONCLUSIONS: Constitutively active RAC1 promotes photoreceptor cell death by oxidative damage that occurs, at least partially, through NOX-induced ROS. Reactive oxygen species are likely involved in multiple forms of retinal degenerations, and our results support investigating RAC1 inhibition as a therapeutic approach that targets this disease pathway.


Subject(s)
Gene Expression Regulation , NADPH Oxidases/metabolism , Neuropeptides/genetics , Oxidative Stress , Retinal Degeneration/metabolism , Retinal Rod Photoreceptor Cells/metabolism , rac1 GTP-Binding Protein/genetics , Animals , Blotting, Western , Cell Death , Disease Models, Animal , Electroretinography , Immunohistochemistry , In Situ Nick-End Labeling , Mice, Transgenic , Neuropeptides/biosynthesis , Polymerase Chain Reaction , RNA/genetics , Retinal Degeneration/genetics , Retinal Degeneration/physiopathology , Retinal Rod Photoreceptor Cells/pathology , rac1 GTP-Binding Protein/biosynthesis
16.
Hum Gene Ther ; 27(5): 376-89, 2016 May.
Article in English | MEDLINE | ID: mdl-27036983

ABSTRACT

Gene therapy for inherited retinal diseases has been shown to ameliorate functional and structural defects in both animal models and in human clinical trials. X-linked retinoschisis (XLRS) is an early-age onset macular dystrophy resulting from loss of an extracellular matrix protein (RS1). In preparation for a human clinical gene therapy trial, we conducted a dose-range efficacy study of the clinical vector, a self-complementary AAV delivering a human retinoschisin (RS1) gene under control of the RS1 promoter and an interphotoreceptor binding protein enhancer (AAV8-scRS/IRBPhRS), in the retinoschisin knockout (Rs1-KO) mouse. The therapeutic vector at 1 × 10(6) to 2.5 × 10(9) (1E6-2.5E9) vector genomes (vg)/eye or vehicle was administered to one eye of 229 male Rs1-KO mice by intravitreal injection at 22 ± 3 days postnatal age (PN). Analysis of retinal function (dark-adapted electroretinogram, ERG), structure (cavities and outer nuclear layer thickness) by in vivo retinal imaging using optical coherence tomography, and retinal immunohistochemistry (IHC) for RS1 was done 3-4 months and/or 6-9 months postinjection (PI). RS1 IHC staining was dose dependent across doses ≥1E7 vg/eye, and the threshold for significant improvement in all measures of retinal structure and function was 1E8 vg/eye. Higher doses, however, did not produce additional improvement. At all doses showing efficacy, RS1 staining in Rs1-KO mouse was less than that in wild-type mice. Improvement in the ERG and RS1 staining was unchanged or greater at 6-9 months than at 3-4 months PI. This study demonstrates that vitreal administration of AAV8 scRS/IRBPhRS produces significant improvement in retinal structure and function in the mouse model of XLRS over a vector dose range that can be extended to a human trial. It indicates that a fully normal level of RS1 expression is not necessary for a therapeutic effect.


Subject(s)
Cell Adhesion Molecules/genetics , Dependovirus/genetics , Eye Proteins/genetics , Genes, X-Linked , Genetic Therapy , Genetic Vectors/genetics , Retinoschisis/genetics , Animals , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Electroretinography , Eye Proteins/metabolism , Gene Expression , Genetic Vectors/administration & dosage , Immunohistochemistry , Intravitreal Injections , Male , Mice , Mice, Knockout , Retina/metabolism , Retina/pathology , Retina/physiopathology , Retinoschisis/diagnosis , Retinoschisis/metabolism , Retinoschisis/therapy , Time Factors , Tomography, Optical Coherence , Transduction, Genetic
17.
Invest Ophthalmol Vis Sci ; 56(11): 6810-22, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26567794

ABSTRACT

PURPOSE: Ciliary neurotrophic factor (CNTF) was recently shown to augment cone function in CNGB3 mutant achromat dogs. However, testing CNTF-releasing implant in human CNGB3 achromats failed to show benefit. We evaluated the effects of CNTF protein on the retinal function in an additional achromatopsia model, the CNGB3-/- mouse. METHODS: Fifty-nine CNGB3-/- mice (postnatal day [PD] ± SD = 30 ± 7) received a unilateral intravitreal injection of 1 or 2 µg CNTF protein, and 15 wild-type (WT) mice (PD = 34 ± 3) received 1 µg CNTF. Retinal function was evaluated by flash ERG and photopic flicker ERG (fERG) at 7 and 14 days after treatment. RESULTS: Seven days post CNTF, the photopic b-wave Vmax was significantly increased in CNGB3-/- mice (P < 0.01), whereas it was reduced in WT mice (P < 0.05). Ciliary neurotrophic factor significantly increased the amplitude of photopic fERG and the photopic oscillatory potentials (OPs) in CNGB3-/- mice. Ciliary neurotrophic factor did not alter the scotopic a-wave in either CNGB3-/- or WT mice, but it increased the scotopic b-wave k (P < 0.01) in CNGB3-/- mice, indicating diminished scotopic sensitivity, and reduced the scotopic b-wave Vmax in WT mice (P < 0.05). No difference was found in ERG parameters between 1 or 2 µg CNTF. Fourteen days after CNTF injection the ERG changes in CNGB3-/- mice were lost. CONCLUSIONS: Intravitreal bolus CNTF protein caused a small and transient improvement of cone-mediated function in CNGB3-/- mice, whereas it reduced rod-mediated function. The increase in photopic OPs and the lack of changes in scotopic a-wave suggest a CNTF effect on the inner retina.


Subject(s)
Ciliary Neurotrophic Factor/administration & dosage , Color Vision Defects/drug therapy , Retinal Cone Photoreceptor Cells/drug effects , Animals , Color Vision Defects/physiopathology , Disease Models, Animal , Drug Implants , Electroretinography , Intravitreal Injections , Mice , Mice, Transgenic , Retinal Cone Photoreceptor Cells/physiology
18.
J Clin Invest ; 125(7): 2891-903, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26098217

ABSTRACT

Strategies aimed at invoking synaptic plasticity have therapeutic potential for several neurological conditions. The human retinal synaptic disease X-linked retinoschisis (XLRS) is characterized by impaired visual signal transmission through the retina and progressive visual acuity loss, and mice lacking retinoschisin (RS1) recapitulate human disease. Here, we demonstrate that restoration of RS1 via retina-specific delivery of adeno-associated virus type 8-RS1 (AAV8-RS1) vector rescues molecular pathology at the photoreceptor-depolarizing bipolar cell (photoreceptor-DBC) synapse and restores function in adult Rs1-KO animals. Initial development of the photoreceptor-DBC synapse was normal in the Rs1-KO retina; however, the metabotropic glutamate receptor 6/transient receptor potential melastatin subfamily M member 1-signaling (mGluR6/TRPM1-signaling) cascade was not properly maintained. Specifically, the TRPM1 channel and G proteins Gαo, Gß5, and RGS11 were progressively lost from postsynaptic DBC dendritic tips, whereas the mGluR6 receptor and RGS7 maintained proper synaptic position. This postsynaptic disruption differed from other murine night-blindness models with an electronegative electroretinogram response, which is also characteristic of murine and human XLRS disease. Upon AAV8-RS1 gene transfer to the retina of adult XLRS mice, TRPM1 and the signaling molecules returned to their proper dendritic tip location, and the DBC resting membrane potential was restored. These findings provide insight into the molecular plasticity of a critical synapse in the visual system and demonstrate potential therapeutic avenues for some diseases involving synaptic pathology.


Subject(s)
Cell Adhesion Molecules/genetics , Eye Proteins/genetics , Genetic Therapy/methods , Retinoschisis/pathology , Retinoschisis/therapy , Animals , Calcium Signaling , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/metabolism , Dependovirus/genetics , Disease Models, Animal , Electroretinography , Eye Proteins/metabolism , Genetic Vectors , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Receptors, Metabotropic Glutamate/metabolism , Retinoschisis/genetics , Synapses/metabolism , Synapses/pathology , TRPM Cation Channels/metabolism
19.
Cold Spring Harb Perspect Med ; 5(8): a017368, 2015 Jun 22.
Article in English | MEDLINE | ID: mdl-26101206

ABSTRACT

Retinoschisis is an X-linked recessive genetic disease that leads to vision loss in males. X-linked retinoschisis (XLRS) typically affects young males; however, progressive vision loss continues throughout life. Although discovered in 1898 by Haas in two brothers, the underlying biology leading to blindness has become apparent only in the last 15 years with the advancement of human genetic analyses, generation of XLRS animal models, and the development of ocular monitoring methods such as the electroretinogram and optical coherence tomography. It is now recognized that retinoschisis results from cyst formations within the retinal layers that interrupt normal visual neurosignaling and compromise structural integrity. Mutations in the human retinoschisin gene have been correlated with disease severity of the human XLRS phenotype. Introduction of a normal human retinoschisin cDNA into retinoschisin knockout mice restores retinal structure and improves neural function, providing proof-of-concept that gene replacement therapy is a plausible treatment for XLRS.


Subject(s)
Genetic Predisposition to Disease , Genetic Therapy/methods , Retinoschisis/genetics , Retinoschisis/therapy , Animals , Disease Models, Animal , Disease Progression , Electroretinography/methods , Female , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/therapy , Genetics, Medical/methods , Humans , Male , Mice , Mice, Knockout , Mutation , Phenotype , Prognosis , Prospective Studies , Rare Diseases , Risk Assessment , Treatment Outcome
20.
J Neurosci ; 34(46): 15356-68, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25392503

ABSTRACT

The mechanisms that specify photoreceptor cell-fate determination, especially as regards to short-wave-sensitive (S) versus medium-wave-sensitive (M) cone identity, and maintain their nature and function, are not fully understood. Here we report the importance of general transcription factor II-I repeat domain-containing protein 1 (GTF2IRD1) in maintaining M cone cell identity and function as well as rod function. In the mouse, GTF2IRD1 is expressed in cell-fate determined photoreceptors at postnatal day 10. GTF2IRD1 binds to enhancer and promoter regions in the mouse rhodopsin, M- and S-opsin genes, but regulates their expression differentially. Through interaction with the transcription factors CRX and thyroid hormone receptor ß 2, it enhances M-opsin expression, whereas it suppresses S-opsin expression; and with CRX and NRL, it enhances rhodopsin expression. In an apparent paradox, although GTF2IRD1 is widely expressed in multiple cell types across the retina, knock-out of GTF2IRD1 alters the retinal expression of only a limited number of annotated genes. Interestingly, however, the null mutation leads to altered topology of cone opsin expression in the retina, with aberrant S-opsin overexpression and M-opsin underexpression in M cones. Gtf2ird1-null mice also demonstrate abnormal M cone and rod electrophysiological responses. These findings suggest an important role for GTF2IRD1 in regulating the level and topology of rod and cone gene expression, and in maintaining normal retinal function.


Subject(s)
Gene Expression Regulation , Muscle Proteins/physiology , Nuclear Proteins/physiology , Retina/metabolism , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Trans-Activators/physiology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Electroretinography , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Opsins/metabolism , Primary Cell Culture , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Rhodopsin/metabolism , Thyroid Hormone Receptors beta/metabolism , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL