Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Am J Pathol ; 193(11): 1706-1720, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36328299

RESUMEN

A pathologic feature of late-onset retinal degeneration caused by the S163R mutation in C1q-tumor necrosis factor-5 (C1QTNF5) is the presence of unusually thick deposits between the retinal pigmented epithelium (RPE) and the vascular choroid, considered a hallmark of this disease. Following its specific expression in mouse RPE, the S163R mutant exhibits a reversed polarized distribution relative to the apically secreted wild-type C1QTNF5, and forms widespread, prominent deposits that gradually increase in size with aging. The current study shows that S163R deposits expand to a considerable thickness through a progressive increase in the basolateral RPE membrane, substantially raising the total RPE height, and enabling their clear imaging as a distinct hyporeflective layer by noninvasive optical coherence tomography in advanced age animals. This phenotype bears a striking resemblance to ocular pathology previously documented in patients harboring the S163R mutation. Therefore, a similar viral vector-based gene delivery approach was used to also investigate the behavior of P188T and G216C, two novel pathogenic C1QTNF5 mutants recently reported in patients for which histopathologic data are lacking. Both mutants primarily impacted the RPE/photoreceptor interface and did not generate basal laminar deposits. Distinct distribution patterns and phenotypic consequences of C1QTNF5 mutants were observed in vivo, which suggested that multiple pathobiological mechanisms contribute to RPE dysfunction and vision loss in this disorder.


Asunto(s)
Degeneración Retiniana , Humanos , Ratones , Animales , Degeneración Retiniana/patología , Mutación , Epitelio Pigmentado de la Retina/metabolismo , Fenotipo
2.
J Biol Chem ; 295(19): 6498-6508, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32238431

RESUMEN

Arrestin-1 is the arrestin family member responsible for inactivation of the G protein-coupled receptor rhodopsin in photoreceptors. Arrestin-1 is also well-known to interact with additional protein partners and to affect other signaling cascades beyond phototransduction. In this study, we investigated one of these alternative arrestin-1 binding partners, the glycolysis enzyme enolase-1, to map the molecular contact sites between these two proteins and investigate how the binding of arrestin-1 affects the catalytic activity of enolase-1. Using fluorescence quench protection of strategically placed fluorophores on the arrestin-1 surface, we observed that arrestin-1 primarily engages enolase-1 along a surface that is opposite of the side of arrestin-1 that binds photoactivated rhodopsin. Using this information, we developed a molecular model of the arrestin-1-enolase-1 complex, which was validated by targeted substitutions of charge-pair interactions. Finally, we identified the likely source of arrestin's modulation of enolase-1 catalysis, showing that selective substitution of two amino acids in arrestin-1 can completely remove its effect on enolase-1 activity while still remaining bound to enolase-1. These findings open up opportunities for examining the functional effects of arrestin-1 on enolase-1 activity in photoreceptors and their surrounding cells.


Asunto(s)
Arrestina/química , Biomarcadores de Tumor/química , Proteínas de Unión al ADN/química , Modelos Moleculares , Complejos Multienzimáticos/química , Fosfopiruvato Hidratasa/química , Rodopsina/química , Proteínas Supresoras de Tumor/química , Arrestina/genética , Sitios de Unión , Biomarcadores de Tumor/genética , Catálisis , Proteínas de Unión al ADN/genética , Humanos , Complejos Multienzimáticos/genética , Fosfopiruvato Hidratasa/genética , Rodopsina/genética , Proteínas Supresoras de Tumor/genética
3.
J Pathol ; 250(2): 195-204, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31625146

RESUMEN

Usher syndrome type 3 (USH3) is an autosomal recessively inherited disorder caused by mutations in the gene clarin-1 (CLRN1), leading to combined progressive hearing loss and retinal degeneration. The cellular distribution of CLRN1 in the retina remains uncertain, either because its expression levels are low or because its epitopes are masked. Indeed, in the adult mouse retina, Clrn1 mRNA is developmentally downregulated, detectable only by RT-PCR. In this study we used the highly sensitive RNAscope in situ hybridization assay and single-cell RNA-sequencing techniques to investigate the distribution of Clrn1 and CLRN1 in mouse and human retina, respectively. We found that Clrn1 transcripts in mouse tissue are localized to the inner retina during postnatal development and in adult stages. The pattern of Clrn1 mRNA cellular expression is similar in both mouse and human adult retina, with CLRN1 transcripts being localized in Müller glia, and not photoreceptors. We generated a novel knock-in mouse with a hemagglutinin (HA) epitope-tagged CLRN1 and showed that CLRN1 is expressed continuously at the protein level in the retina. Following enzymatic deglycosylation and immunoblotting analysis, we detected a single CLRN1-specific protein band in homogenates of mouse and human retina, consistent in size with the main CLRN1 isoform. Taken together, our results implicate Müller glia in USH3 pathology, placing this cell type to the center of future mechanistic and therapeutic studies to prevent vision loss in this disease. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Células Ependimogliales/metabolismo , Proteínas de la Membrana/biosíntesis , Retina/metabolismo , Síndromes de Usher/metabolismo , Animales , Glicosilación , Humanos , Hibridación in Situ , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Neuroglía/metabolismo , ARN Mensajero/genética , Síndromes de Usher/patología
4.
Ophthalmic Res ; 64(1): 91-98, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32535604

RESUMEN

INTRODUCTION: Bevacizumab and ranibizumab, which are anti-vascular endothelial growth factor (VEGF) medications, are used frequently in the treatment for retinopathy of prematurity (ROP) in infants. Aflibercept, or VEGF Trap, has been used anecdotally, but translation and clinical studies are lacking. OBJECTIVE: This study investigates the efficacy of aflibercept at reducing areas of non-perfused retina and studies its effect on normal angiogenesis in the oxygen-induced retinopathy mouse model of ROP. METHODS: C57BL/6 J mice were assigned to room air control (n = 21 eyes) or hyperoxia with 75% oxygen (n = 84 eyes). The hyperoxic mice were assigned to 1 of 3 groups: 0 ng (n = 14 eyes), 100 ng (n = 35 eyes), or 1,000 ng (n = 35 eyes) of intravitreal aflibercept administered on postnatal day 14. Eyes were enucleated at PN17 and PN25 postinjection. Retinas were stained with anti-collagen IV antibody and photographed with microscopy. Areas of perfused and non-perfused retina were quantified using ImageJ software. Statistical comparisons were made using ANOVA with Tukey post hoc comparisons. RESULTS: At PN17, there was no significant difference in the area of non-perfused retina between the hyperoxic control and the 100 and 1,000 ng aflibercept groups. At PN25, the 100 ng (p < 0.05) and 1,000 ng (p = 0.008) treatment groups displayed less non-perfusion compared to hyperoxic controls. At the 1,000 ng dose, there was increased non-perfusion compared to the 100 ng dose (p = 0.02). There was reduced non-perfusion by PN25 compared to PN17 for the 100 ng group (p < 0.05), with no difference in the 1,000 ng group. CONCLUSIONS: The study shows that the area of non-perfused retina decreases effectively with aflibercept at PN25 with 100 ng dosage. With the 1,000 ng dosage, there is an inhibition of the physiologic angiogenesis with a higher area of non-perfused retina.


Asunto(s)
Receptores de Factores de Crecimiento Endotelial Vascular/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Retina/fisiopatología , Retinopatía de la Prematuridad/tratamiento farmacológico , Agudeza Visual , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Modelos Animales de Enfermedad , Inyecciones Intravítreas , Ratones , Ratones Endogámicos C57BL , Oxígeno/toxicidad , Retina/efectos de los fármacos , Retinopatía de la Prematuridad/inducido químicamente , Retinopatía de la Prematuridad/fisiopatología , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
5.
Mol Vis ; 24: 17-28, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29386880

RESUMEN

Purpose: Blue cone monochromacy (BCM) is an X-linked congenital vision disorder characterized by complete loss or severely reduced L- and M-cone function. Patients with BCM display poor visual acuity, severely impaired color discrimination, myopia, nystagmus, and minimally detectable cone-mediated electroretinogram. Recent studies of patients with BCM with adaptive optics scanning laser ophthalmoscopy (AOSLO) showed that they have a disrupted cone mosaic with reduced numbers of cones in the fovea that is normally dominated by L- and M-cones. The remaining cones in the fovea have significantly shortened outer segments but retain sufficient structural integrity to serve as potential gene therapy targets. In this study, we tested whether exogenously expressed human L- and M-opsins can rescue M-cone function in an M-opsin knockout (Opn1mw-/- ) mouse model for BCM. Methods: Adeno-associated virus type 5 (AAV5) vectors expressing OPN1LW, OPN1MW, or C-terminal tagged OPN1LW-Myc, or OPN1MW-HA driven by a cone-specific promoter were injected subretinally into one eye of Opn1mw-/- mice, while the contralateral eye served as the uninjected control. Expression of cone pigments was determined with western blotting and their cellular localization identified with immunohistochemistry. M-cone function was analyzed with electroretinogram (ERG). Antibodies against cone phototransduction proteins were used to study cone outer segment (OS) morphology in untreated and treated Opn1mw-/- eyes. Results: We showed that cones in the dorsal retina of the Opn1mw-/- mouse do not form outer segments, resembling cones that lack outer segments in the human BCM fovea. We further showed that AAV5-mediated expression of either human M- or L-opsin individually or combined promotes regrowth of cone outer segments and rescues M-cone function in the treated Opn1mw-/- dorsal retina. Conclusions: Exogenously expressed human opsins can regenerate cone outer segments and rescue M-cone function in Opn1mw-/- mice, thus providing a proof-of-concept gene therapy in an animal model of BCM.


Asunto(s)
Defectos de la Visión Cromática/terapia , Fóvea Central/metabolismo , Terapia Genética/métodos , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Opsinas de Bastones/genética , Animales , Defectos de la Visión Cromática/genética , Defectos de la Visión Cromática/metabolismo , Defectos de la Visión Cromática/patología , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Fóvea Central/patología , Expresión Génica , Prueba de Complementación Genética , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Ratones , Ratones Noqueados , Oftalmoscopía , Regiones Promotoras Genéticas , Segmento Externo de las Células Fotorreceptoras Retinianas/patología , Opsinas de Bastones/metabolismo , Transgenes
6.
Adv Exp Med Biol ; 1074: 61-66, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721928

RESUMEN

The pathogenic mutation S163R in C1QTNF5 causes a disorder known as autosomal dominant late-onset retinal degeneration (L-ORD), characterized by the presence of thick extracellular sub-RPE deposits, similar histopathologically to those found in AMD patients. We have previously shown that the S163R C1QTNF5 mutant forms globular aggregates within the RPE in vivo following its AAV-mediated expression in the RPE and exhibits a reversely polarized distribution, being routed toward the basal rather than apical RPE. We show here that when both wild-type and mutant S163R C1QTNF5 are simultaneously delivered subretinally to mouse RPE cells, the mutant impairs the wild-type protein secretion from the RPE, and both proteins are dispersed toward the basal and lateral RPE membrane. This result has mechanistic and therapeutic implications for L-ORD disorder.


Asunto(s)
Degeneración Macular/genética , Mutación Missense , Mutación Puntual , Agregación Patológica de Proteínas/genética , Epitelio Pigmentado de la Retina/metabolismo , Animales , Polaridad Celular , Colágeno/química , Colágeno/genética , Colágeno/metabolismo , Dependovirus/genética , Electrorretinografía , Genes Dominantes , Vectores Genéticos , Humanos , Inyecciones Intraoculares , Degeneración Macular/metabolismo , Degeneración Macular/patología , Ratones , Ratones Endogámicos C57BL , Agregación Patológica de Proteínas/patología , Proteínas Recombinantes/análisis , Proteínas Recombinantes/metabolismo , Epitelio Pigmentado de la Retina/ultraestructura , Fracciones Subcelulares/química
7.
Int J Mol Sci ; 18(4)2017 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-28353645

RESUMEN

The accumulation of lipofuscin in the retinal pigment epithelium (RPE) is dependent on the effectiveness of photoreceptor outer segment material degradation. This study explored the role of autophagy in the fate of RPE lipofuscin degradation. After seven days of feeding with either native or modified rod outer segments, ARPE-19 cells were treated with enhancers or inhibitors of autophagy and the autofluorescence was detected by fluorescence-activated cell sorting. Supplementation with different types of rod outer segments increased lipofuscin-like autofluorescence (LLAF) after the inhibition of autophagy, while the induction of autophagy (e.g., application of rapamycin) decreased LLAF. The effects of autophagy induction were further confirmed by Western blotting, which showed the conversion of LC3-I to LC3-II, and by immunofluorescence microscopy, which detected the lysosomal activity of the autophagy inducers. We also monitored LLAF after the application of several autophagy inhibitors by RNA-interference and confocal microscopy. The results showed that, in general, the inhibition of the autophagy-related proteins resulted in an increase in LLAF when cells were fed with rod outer segments, which further confirms the effect of autophagy in the fate of RPE lipofuscin degradation. These results emphasize the complex role of autophagy in modulating RPE autofluorescence and confirm the possibility of the pharmacological clearance of RPE lipofuscin by small molecules.


Asunto(s)
Autofagia , Lipofuscina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Línea Celular , Fluorescencia , Humanos , Lisosomas/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Sirolimus/farmacología
8.
Cell Mol Life Sci ; 70(23): 4603-16, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23817741

RESUMEN

Arrestins are dynamic proteins that move between cell compartments triggered by stimulation of G-protein-coupled receptors. Even more dynamically in vertebrate photoreceptors, arrestin1 (Arr1) moves between the inner and outer segments according to the light conditions. Previous studies have shown that the light-driven translocation of Arr1 in rod photoreceptors is initiated by rhodopsin through a phospholipase C/protein kinase C (PKC) signaling cascade. The purpose of this study is to identify the PKC substrate that regulates the translocation of Arr1. Mass spectrometry was used to identify the primary phosphorylated proteins in extracts prepared from PKC-stimulated mouse eye cups, confirming the finding with in vitro phosphorylation assays. Our results show that Bardet-Biedl syndrome 5 (BBS5) is the principal protein phosphorylated either by phorbol ester stimulation or by light stimulation of PKC. Via immunoprecipitation of BBS5 in rod outer segments, Arr1 was pulled down; phosphorylation of BBS5 reduced this co-precipitation of Arr1. Immunofluorescence and immunoelectron microscopy showed that BBS5 principally localizes along the axonemes of rods and cones, but also in photoreceptor inner segments, and synaptic regions. Our principal findings in this study are threefold. First, we demonstrate that BBS5 is post-translationally regulated by phosphorylation via PKC, an event that is triggered by light in photoreceptor cells. Second, we find a direct interaction between BBS5 and Arr1, an interaction that is modulated by phosphorylation of BBS5. Finally, we show that BBS5 is distributed along the photoreceptor axoneme, co-localizing with Arr1 in the dark. These findings suggest a role for BBS5 in regulating light-dependent translocation of Arr1 and a model describing its role in Arr1 translocation is proposed.


Asunto(s)
Arrestinas/metabolismo , Proteínas Portadoras/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , Animales Modificados Genéticamente , Arrestinas/genética , Axonema/metabolismo , Proteínas Portadoras/genética , Proteínas del Citoesqueleto , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Immunoblotting , Luz , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Inmunoelectrónica , Modelos Biológicos , Proteínas de Unión a Fosfato , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación/efectos de la radiación , Unión Proteica/efectos de la radiación , Proteína Quinasa C/metabolismo , Células Fotorreceptoras Retinianas Bastones/ultraestructura , Xenopus , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
9.
Mol Vis ; 19: 2579-89, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-24367157

RESUMEN

Mouse models are useful tools for developing potential therapies for human inherited retinal diseases, such as retinitis pigmentosa (RP), since more strains are being identified with the same mutant genes and phenotypes as humans with corresponding retinal degenerative diseases. Mutations in the beta subunit of the human rod phosphodiesterase (PDE6B) gene are a common cause of autosomal recessive RP (arRP). This article focuses on two well-established naturally occurring mouse models of arRP caused by spontaneous mutations in Pde6b, their discovery, phenotype, mechanism of degeneration, strengths and limitations, and therapeutic approaches to restore vision and delay disease progression. Viral vector, especially adeno-associated viral vector (AAV) -mediated gene replacement therapy, pharmacological treatment, cell-based therapy and other approaches that extend the therapeutic window of treatment, is a potentially promising strategy for improving photoreceptor function and significantly slowing the process of retinal degeneration.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Mutación , Retina/enzimología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Adenoviridae/genética , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Modelos Animales de Enfermedad , Expresión Génica , Terapia Genética/métodos , Vectores Genéticos , Humanos , Ratones , Retina/patología , Retinitis Pigmentosa/enzimología , Retinitis Pigmentosa/patología
10.
Artículo en Inglés | MEDLINE | ID: mdl-35940643

RESUMEN

Mutations in RHO, the gene for rhodopsin, account for a large fraction of autosomal-dominant retinitis pigmentosa (adRP). Patients fall into two clinical classes, those with early onset, pan retinal photoreceptor degeneration, and those who experience slowly progressive disease. The latter class of patients are candidates for photoreceptor-directed gene therapy, while former may be candidates for delivery of light-responsive proteins to interneurons or retinal ganglion cells. Gene therapy for RHO adRP may be targeted to the mutant gene at the DNA or RNA level, while other therapies preserve the viability of photoreceptors without addressing the underlying mutation. Correcting the RHO gene and replacing the mutant RNA show promise in animal models, while sustaining viable photoreceptors has the potential to delay the loss of central vision and may preserve photoreceptors for gene-directed treatments.

11.
Hum Gene Ther ; 33(13-14): 695-707, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35081746

RESUMEN

Glucose metabolism in the retina is carefully orchestrated, with glucose being delivered to photoreceptors from the choroidal circulation through the retinal pigmented epithelium (RPE). In photoreceptors, glucose is processed principally by aerobic glycolysis, from which the lactate byproduct is provided to the RPE and Müller glia for their energetic needs. In this study, we utilize a modified arrestin1 protein to enhance the glycolytic output of lactate from rod photoreceptors through disinhibition of enolase1 activity with the goal being to use this increased lactate production as a gene-agnostic approach to slowing retinal degeneration. Mouse arrestin1 with E362G/D363G amino acid substitutions (referred to as "ArrGG") was packaged into AAV and tested for safety and for efficacy in increasing retinal lactate production. Overexpression of ArrGG in C57BL/6J mice did not result in any detectable changes in either electroretinogram (ERG) function or photoreceptor survival as measured by outer nuclear layer (ONL) thickness. However, mouse retinas expressing ArrGG showed a ∼25% increase in the rate of lactate secretion. Therefore, AAV-ArrGG was delivered intravitreally to heterozygous P23H rhodopsin knockin mice (RhoP23H/+) to determine if enhancing glycolysis in photoreceptors can slow retinal degeneration in this animal model of retinitis pigmentosa. We found that the expression of ArrGG in these mice slowed the decline of both scotopic and photopic ERG function. Correspondingly, there was significant preservation of ONL thickness in RhoP23H/+ mice treated with ArrGG compared with controls. In conclusion, our studies show that expressing ArrGG in C57BL/6J mouse retina results in an increase in lactate production, consistent with an upregulation of glycolysis. In the P23H rhodopsin model of retinitis pigmentosa, the expression of ArrGG led to significant preservation of photoreceptor function and slowing of retinal degeneration. These findings suggest that enhancing glycolysis by targeting increased enolase1 activity with a modified arrestin1 in photoreceptors may offer a therapeutic approach to slowing retinal degeneration.


Asunto(s)
Degeneración Retiniana , Retinitis Pigmentosa , Animales , Arrestinas , Modelos Animales de Enfermedad , Electrorretinografía , Glucosa , Ácido Láctico , Ratones , Ratones Endogámicos C57BL , Retina/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/terapia , Retinitis Pigmentosa/terapia , Rodopsina/genética
12.
Transl Vis Sci Technol ; 11(11): 8, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36355386

RESUMEN

Purpose: Ocular hypertension is a significant risk factor for vision loss in glaucoma caused by the death of retinal ganglion cells (RGCs). We investigated whether small heat shock proteins (sHsps) expressed in RGCs protect those cells against ocular hypertension in mice. Methods: AAV2 vectors encoding genes for one of the following four human sHsps: HSPB1, HSPB4, HSPB5, or HSPB6 were constructed for RGC-specific expression. Ischemia/reperfusion was induced by elevating the intraocular pressure (IOP) to 120 mm Hg for one hour, followed by a rapid return to normal IOP. Microbeads (MB) were injected into the anterior chamber of mice to induce ocular hypertension. RGC death and glial activation were assessed by immunostaining for Brn3a, RBPMS, Iba1, and glial fibrillary acid protein in retinal flat mounts. RGC axonal defects were evaluated by anterograde transport of intravitreally injected cholera toxin-B. RGC function was assessed by pattern electroretinography. Results: Among the sHsps, HspB1 offered the best protection against RGC death from ischemia/reperfusion injury in the mouse retina. Intravitreal administration of AAV2-HSPB1 either two weeks before or one week after instituting ocular hypertension resulted in significant prevention of RGC loss. The MB-injected mice showed RGC axonal transportation defects, but AAV2-HSPB1 administration significantly inhibited this defect. AAV2-HSPB1 prevented glial activation caused by ocular hypertension. More importantly, a single injection of AAV2-HSPB1 protected RGCs long-term in MB-injected eyes. Conclusions: The administration of AAV2-HSPB1 inhibited RGC death and axonal transport defects and reduced glial activation in a mouse model of ocular hypertension. Translational Relevance: Our results suggested that the intravitreal delivery of AAV2-HSPB1 could be developed as a gene therapy to prevent vision loss on a long-term basis in glaucoma patients.


Asunto(s)
Glaucoma , Hipertensión Ocular , Humanos , Ratones , Animales , Células Ganglionares de la Retina/metabolismo , Transporte Axonal , Hipertensión Ocular/genética , Hipertensión Ocular/metabolismo , Glaucoma/genética , Glaucoma/prevención & control , Presión Intraocular , Modelos Animales de Enfermedad , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo
13.
PLoS One ; 17(5): e0269261, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35639753

RESUMEN

Glaucoma is the leading cause of irreversible blindness worldwide. Therapies for glaucoma are directed toward reducing intraocular pressure (IOP), the leading risk factor and only reliable therapeutic target via topical medications or with procedural intervention including laser or surgery. Though topical therapeutics are typically first line, less than 50% of patients take drops as prescribed. Sustained release technologies that decrease IOP for extended periods of time are being examined for clinical use. We recently identified Stanniocalcin-1, a naturally occurring hormone, as an IOP-lowering agent. Here, we show that a single injection into the anterior chamber of mice with an adeno-associated viral vector containing the transgene of stanniocalcin-1 results in diffuse and sustained expression of the protein and produces IOP reduction for up to 6 months. As the treatment effect begins to wane, IOP-lowering can be rescued with a repeat injection. Aqueous humor dynamic studies revealed an increase in outflow facility as the mechanism of action. This first-in-class therapeutic approach has the potential to improve care and reduce the rates of vision loss in the 80 million people worldwide currently affected by glaucoma.


Asunto(s)
Glaucoma , Hipotensión Ocular , Animales , Glaucoma/tratamiento farmacológico , Glaucoma/genética , Glicoproteínas , Humanos , Presión Intraocular , Ratones , Tonometría Ocular , Transgenes
14.
Mol Vis ; 16: 916-34, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-20517486

RESUMEN

PURPOSE: Growing evidence suggests that successful treatment of many inherited photoreceptor diseases will require multi-protein therapies that not only correct the genetic defects linked to these diseases but also slow or halt the related degenerative phenotypes. To be effective, it is likely that therapeutic protein expression will need to be targeted to specific cell types. The purpose of this study was to develop dual-promoter lentiviral vectors that target expression of two proteins to retinal cones and rods, rods only, or Müller cells. METHODS: Dual-promoter lentivectors were constructed using the following promoters: Xenopus opsin promoter (XOPS)1.3, murine opsin promoter (MOPS), interphotoreceptor retinoid binding protein promoter (IRBP156), rhodopsin kinase (RK), neural retina leucine zipper (NRLL), vimentin (VIM), cluster differentiation (CD44), and glial fibrillary acidic protein (GFAP). Vectors were packaged and injected into the neural tubes of chicken embryos. The activities of the promoters alone, in duplicate, or when paired with a different promoter were analyzed in transduced, fully-developed retinas, using direct fluorescent and immunofluorescent microscopy. RESULTS: IRBP156, NRLL, and RK were active in cones and rods while XOPS1.3 was active only in rods. Of the glial promoters, only GFAP activity was restricted to Müller cells; both VIM and CD44 were active in Müller and neural cells. Dual-promoter vectors carrying IRBP156 and RK or XOPS1.3 and MOPS, in the order listed, exhibited robust expression of both reporter transgenes in cones and rods or rods only, respectively. Expression of the upstream transgene was much lower than the downstream transgene in dual-promoter vectors constructed using two copies of either RK or IRBP156. Analyses of the expression of a dual-promoter vector carrying CD44 and VIM in the order listed showed that the activity of the VIM promoter was more restricted to glial cells when paired with the CD44 promoter, while the activity of the CD44 promoter was inhibited to the extent that no CD44-driven reporter protein was detected in transduced cells. CONCLUSIONS: We have identified two dual-promoter vectors, one that targets cones and rods and one that targets rods alone. Both vectors reliably express the two proteins encoded by the transgenes they carry. When two well matched promoters are not available, we found that it is possible to target expression of two proteins to single cells using dual-promoter vectors carrying two copies of the same promoter. These vectors should be useful in studies of retina when co-delivery of a reporter protein with an experimental protein is desired or when expression of two exogenous proteins in targeted cells is required.


Asunto(s)
Marcación de Gen/métodos , Vectores Genéticos , Lentivirus/genética , Células Fotorreceptoras de Vertebrados , Regiones Promotoras Genéticas , Retina/citología , Animales , Embrión de Pollo , Expresión Génica , Ratones/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/embriología , Retina/metabolismo , Transgenes , Xenopus/genética
15.
Invest Ophthalmol Vis Sci ; 60(10): 3644-3651, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31469404

RESUMEN

Purpose: Previously we showed that AAV5-mediated expression of either human M- or L-opsin promoted regrowth of cone outer segments and rescued M-cone function in the treated M-opsin knockout (Opn1mw-/-) dorsal retina. In this study, we determined cone viability and window of treatability in aged Opn1mw-/- mice. Methods: Cone viability was assessed with antibody against cone arrestin and peanut agglutinin (PNA) staining. The rate of cone degeneration in Opn1mw-/- mice was quantified by PNA staining. AAV5 vector expressing human L-opsin was injected subretinally into one eye of Opn1mw-/- mice at 1, 7, and 15 months old, while the contralateral eyes served as controls. M-cone-mediated retinal function was analyzed 2 and 13 months postinjection by full-field ERG. L-opsin transgene expression and cone outer segment structure were examined by immunohistochemistry. Results: We showed that dorsal M-opsin dominant cones exhibit outer segment degeneration at an early age in Opn1mw-/- mice, whereas ventral S-opsin dominant cones were normal. The remaining M-opsin dominant cones remained viable for at least 15 months, albeit having shortened or no outer segments. We also showed that AAV5-mediated expression of human L-opsin was still able to rescue function and outer segment structure in the remaining M-opsin dominant cones when treatment was initiated at 15 months of age. Conclusions: Our results showing that the remaining M-opsin dominant cones in aged Opn1mw-/- mice can still be rescued by gene therapy is helpful for establishing the window of treatability in future blue cone monochromacy clinical trials.


Asunto(s)
Defectos de la Visión Cromática/terapia , Terapia Genética/métodos , Células Fotorreceptoras Retinianas Conos/fisiología , Opsinas de Bastones/genética , Opsinas de Bastones/fisiología , Envejecimiento/fisiología , Animales , Arrestinas/genética , Defectos de la Visión Cromática/genética , Defectos de la Visión Cromática/fisiopatología , Dependovirus , Modelos Animales de Enfermedad , Electrorretinografía , Regulación de la Expresión Génica/fisiología , Vectores Genéticos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Parvovirinae/genética , Retina/fisiopatología
16.
Mol Vis ; 14: 615-24, 2008 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-18385797

RESUMEN

PURPOSE: Anterior chamber-associated immune deviation (ACAID) is a form of peripheral tolerance achieved via intracameral antigen inoculation. It is well known that ACAID effectively down-regulates potentially destructive immunities such as delayed-type hypersensitivity (DTH) at extraorbital sites. However, what has not been specifically addressed is whether local intraocular tissues are negatively affected from intracamerally placed antigen. Thus, the current study was undertaken to detect and characterize potential pathological effects on intraocular tissues following intracameral inoculation with alloantigen. METHODS: ACAID induced in C57BL/6 hosts via intracameral inoculation with allogeneic (BALB/c) splenocytes was confirmed by the absence of DTH reactivity in the periphery. Injuries to the anterior segment and neuroretina following intracameral inoculation were evaluated pathologically via histological evaluation, molecularly via upregulation of glial fibrillary acidic protein (GFAP), and functionally via assessment of photoreceptor degeneration and electroretinogram (ERG) out to 24 days. In all experiments, intracamerally inoculated mice were compared to sham-operated, and controlled lens-punctured mice--a procedure that elicits intracameral inflammation for positive identification of immunopathological changes. RESULTS: Inflammation of anterior segment tissues persisted out to eight days, despite evidence that significant clearance of allogeneic cells took place within 6 h. In the neuroretina, a transient loss in ERG B-wave amplitudes was detected, but photoreceptor degeneration and GFAP upregulation were not. CONCLUSIONS: Intracameral inoculation with alloantigen leads to anterior segment inflammation and ERG dysfunction; however, this was markedly reduced and transient when compared to strong anterior segment inflammation induced by a more serious lens-puncture wound.


Asunto(s)
Cámara Anterior/inmunología , Ojo/inmunología , Ojo/patología , Tolerancia Inmunológica , Isoantígenos/inmunología , Animales , Segmento Anterior del Ojo/patología , Electrorretinografía , Gliosis/inmunología , Gliosis/patología , Inflamación/inmunología , Inflamación/patología , Inyecciones , Isoantígenos/administración & dosificación , Cristalino/lesiones , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Fotorreceptoras/patología , Retina/inmunología , Retina/fisiopatología , Bazo/inmunología , Factores de Tiempo , Heridas Penetrantes/patología
17.
J Clin Invest ; 111(8): 1171-80, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12697736

RESUMEN

Experimental autoimmune uveitis (EAU) is a disease of the neural retina induced by immunization with retinal antigens, such as interphotoreceptor retinoid-binding protein (IRBP) and arrestin (retinal soluble antigen, S-Ag). EAU serves as a model for human autoimmune uveitic diseases associated with major histocompatibility complex (HLA) genes, in which patients exhibit immunological responses to retinal antigens. Here we report the development of a humanized EAU model in HLA transgenic (TG) mice. HLA-DR3, -DR4, -DQ6, and -DQ8 TG mice were susceptible to IRBP-induced EAU. Importantly, HLA-DR3 TG mice developed severe EAU with S-Ag, to which wild-type mice are highly resistant. Lymphocyte proliferation was blocked by anti-HLA antibodies, confirming that antigen is functionally presented by the human MHC molecules. Disease could be transferred by immune cells with a Th1-like cytokine profile. Antigen-specific T cell repertoire, as manifested by responses to overlapping peptides derived from S-Ag or IRBP, differed from that of wild-type mice. Interestingly, DR3 TG mice, but not wild-type mice, recognized an immunodominant S-Ag epitope between residues 291 and 310 that overlaps with a region of S-Ag recognized by uveitis patients. Thus, EAU in HLA TG mice offers a new model of uveitis that should represent human disease more faithfully than currently existing models.


Asunto(s)
Enfermedades Autoinmunes/etiología , Proteínas del Ojo , Antígenos de Histocompatibilidad Clase II/fisiología , Uveítis/etiología , Secuencia de Aminoácidos , Animales , Presentación de Antígeno , Arrestina/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T , Antígeno HLA-DR3/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas de Unión al Retinol/inmunología
18.
Mol Vis ; 12: 1516-25, 2006 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-17167410

RESUMEN

PURPOSE: The purpose of our study was to determine whether arrestin residues previously predicted by computational modeling to interact with an aspartic acid substituted rhodopsin tail are actually involved in interactions with phospho-residues on the rhodopsin cytoplasmic tail. METHODS: We generated arrestin mutants with altered charges at predicted positions. These mutants were then tested for the ability to inhibit rhodopsin using both direct binding assays, as well as functional assays involving transducin inhibition assays. RESULTS: Our results demonstrate that the computer-predicted residues are indeed involved in both the ability of the low-affinity state of arrestin to bind to rhodopsin as well as the ability of arrestin to be induced into a higher-affinity state in a phospho-residue-dependent manner. CONCLUSIONS: Our results also suggest that positions K14, K15, R29, H301, and K300 on arrestin interact with the phosphorylated carboxyl tail of rhodopsin and that this translates to the efficient activation of arrestin.


Asunto(s)
Arrestina/metabolismo , Rodopsina/metabolismo , Animales , Arrestina/efectos de los fármacos , Arrestina/genética , Unión Competitiva , Bovinos , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Fotólisis , Rodopsina/química , Segmento Externo de la Célula en Bastón/metabolismo , Tripsina/farmacología
19.
Vision Res ; 46(27): 4464-71, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16979692

RESUMEN

Vertebrate photoreceptor cells are polarized neurons highly specialized for light absorption and visual signal transduction. Photoreceptor cells consist of the light sensitive outer segment and the biosynthetic active inner segment linked by a slender connecting cilium. The function of mature photoreceptor cells is strictly dependent on this compartmentalization which is maintained in the specialized retinal environment. To keep this fragile morphologic and functional composition for further cell biological studies and treatments we established organotypic retina cultures of mature mice and Xenopus laevis. The organotypic retina cultures of both model organisms are created as co-cultures of the retina and the pigment epithelium, still attached to outer segments of the photoreceptor cells. To demonstrate the suitability of the culture system for physiological analyses we performed apoptotic cell death analyses and verified photoreceptor viability. Furthermore, light-dependent bidirectional movements of arrestin and transducin in photoreceptors in vivo and in the retinal cultures were indistinguishable indicating normal photoreceptor cell-biologic function in organotypic cultures. Our established culture systems allow the analysis of mature photoreceptor cells and their accessibility to treatments, characteristic for common cell culture. Furthermore, this culturing technique also provides an appropriate system for gene delivery to retinal cells and will serve to simulate gene therapeutic approaches prior to difficult and time-consuming in vivo experiments.


Asunto(s)
Células Fotorreceptoras/fisiología , Retina/metabolismo , Vertebrados/metabolismo , Animales , Arrestina/metabolismo , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Técnicas de Cultivo de Órganos , Estimulación Luminosa , Transporte de Proteínas , Retina/anatomía & histología , Transducina/metabolismo , Visión Ocular/fisiología , Xenopus
20.
PLoS One ; 11(2): e0148773, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26867008

RESUMEN

PURPOSE: Bardet-Biedl syndrome is a complex ciliopathy that usually manifests with some form of retinal degeneration, amongst other ciliary-related deficiencies. One of the genetic causes of this syndrome results from a defect in Bardet-Biedl Syndrome 5 (BBS5) protein. BBS5 is one component of the BBSome, a complex of proteins that regulates the protein composition in cilia. In this study, we identify a smaller molecular mass form of BBS5 as a variant formed by alternative splicing and show that expression of this splice variant is restricted to the retina. METHODS: Reverse transcription PCR from RNA was used to isolate and identify potential alternative transcripts of Bbs5. A peptide unique to the C-terminus of the BBS5 splice variant was synthesized and used to prepare antibodies that selectively recognized the BBS5 splice variant. These antibodies were used on immunoblots of tissue extracts to determine the extent of expression of the alternative transcript and on tissue slices to determine the localization of expressed protein. Pull-down of fluorescently labeled arrestin1 by immunoprecipitation of the BBS5 splice variant was performed to assess functional interaction between the two proteins. RESULTS: PCR from mouse retinal cDNA using Bbs5-specific primers amplified a unique cDNA that was shown to be a splice variant of BBS5 resulting from the use of cryptic splicing sites in Intron 7. The resulting transcript codes for a truncated form of the BBS5 protein with a unique 24 amino acid C-terminus, and predicted 26.5 kD molecular mass. PCR screening of RNA isolated from various ciliated tissues and immunoblots of protein extracts from these same tissues showed that this splice variant was expressed in retina, but not brain, heart, kidney, or testes. Quantitative PCR showed that the splice variant transcript is 8.9-fold (+/- 1.1-fold) less abundant than the full-length transcript. In the retina, the splice variant of BBS5 appears to be most abundant in the connecting cilium of photoreceptors, where BBS5 is also localized. Like BBS5, the binding of BBS5L to arrestin1 can be modulated by phosphorylation through protein kinase C. CONCLUSIONS: In this study we have identified a novel splice variant of BBS5 that appears to be expressed only in the retina. The BBS5 splice variant is expressed at approximately 10% of full-length BBS5 level. No unique functional or localization properties could be identified for the splice variant compared to BBS5.


Asunto(s)
Empalme Alternativo , Síndrome de Bardet-Biedl/genética , Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Proteínas/metabolismo , Retina/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/genética , Bovinos , Cilios/metabolismo , Proteínas del Citoesqueleto , ADN Complementario/metabolismo , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neuronas/metabolismo , Proteínas de Unión a Fosfato , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Proteínas/genética , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sus scrofa , Distribución Tisular , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA