Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 161(4): 817-32, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25957687

RESUMEN

Rod-derived cone viability factor (RdCVF) is an inactive thioredoxin secreted by rod photoreceptors that protects cones from degeneration. Because the secondary loss of cones in retinitis pigmentosa (RP) leads to blindness, the administration of RdCVF is a promising therapy for this untreatable neurodegenerative disease. Here, we investigated the mechanism underlying the protective role of RdCVF in RP. We show that RdCVF acts through binding to Basigin-1 (BSG1), a transmembrane protein expressed specifically by photoreceptors. BSG1 binds to the glucose transporter GLUT1, resulting in increased glucose entry into cones. Increased glucose promotes cone survival by stimulation of aerobic glycolysis. Moreover, a missense mutation of RdCVF results in its inability to bind to BSG1, stimulate glucose uptake, and prevent secondary cone death in a model of RP. Our data uncover an entirely novel mechanism of neuroprotection through the stimulation of glucose metabolism.


Asunto(s)
Proteínas del Ojo/metabolismo , Glucólisis , Tiorredoxinas/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Basigina/genética , Basigina/metabolismo , Proteínas del Ojo/genética , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Ratones , Mutación Missense , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/metabolismo , Retinitis Pigmentosa/metabolismo , Tiorredoxinas/genética
2.
Adv Exp Med Biol ; 1415: 117-123, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37440023

RESUMEN

Gene therapy is a potential cure for several inherited retinal dystrophies, and adeno-associated virus (AAV) has emerged as a vector of choice for therapeutic gene delivery to the retina. However, prior exposure to AAVs can cause a humoral immune response resulting in the presence of antibodies in the serum, which can subsequently interfere with the AAV-mediated gene therapy. The antibodies bind specifically to a serotype but often display broad cross-reactivity. A subset of these antibodies called neutralizing antibodies (NABs) can render the AAV inactive, thereby reducing the efficacy of the therapy. The preexisting NAB levels against different serotypes vary by species, and these variations need to be considered while designing studies. Since large animals often serve as preclinical models to test gene therapies, in this review we compile studies reporting preexisting NABs against commonly used AAV serotypes in humans and large animal models and discuss strategies to deal with NABs.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Animales , Humanos , Dependovirus/genética , Serogrupo , Vectores Genéticos/genética , Terapia Genética/métodos , Modelos Animales
3.
Ophthalmic Res ; 2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36103843

RESUMEN

The recent approval of voretigene neparvovec (Luxturna®) for patients with biallelic RPE65 mutation-associated inherited retinal dystrophy with viable retinal cells represents an important step in the development of ocular gene therapies. Herein, we review studies investigating the episomal persistence of different recombinant adeno-associated virus (rAAV) vector genomes and the pre-clinical and clinical evidence of long-term effects of different RPE65 gene replacement therapies. A targeted review of articles published between 1974 and January 2021 in Medline®, Embase®, and other databases, was conducted, followed by a descriptive longitudinal analysis of the clinical trial outcomes of voretigene neparvovec. Following an initial screening, 14 publications examining the episomal persistence of different rAAV genomes and 71 publications evaluating gene therapies in animal models were included. Viral genomes were found to persist for at least 22 months (longest study follow-up) as transcriptionally active episomes. Treatment effects lasting almost a decade were reported in canine disease models, with more pronounced effects the earlier the intervention. The clinical trial outcomes of voretigene neparvovec are consistent with pre-clinical findings and reveal sustained results for up to 7.5 years for the full-field light sensitivity threshold test and 5 years for the multi-luminance mobility test in the Phase I and Phase III trials, respectively. In conclusion, the therapeutic effect of voretigene neparvovec lasts for at least a decade in animal models and 7.5 years in human subjects. Since retinal cells can retain functionality over their lifetime after transduction, these effects may be expected to last even longer in patients with a sufficient number of outer retinal cells at the time of intervention.

4.
Proc Natl Acad Sci U S A ; 116(52): 26280-26287, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31871177

RESUMEN

Retinal degenerative diseases caused by photoreceptor cell death are major causes of irreversible vision loss. As only primates have a macula, the nonhuman primate (NHP) models have a crucial role not only in revealing biological mechanisms underlying high-acuity vision but also in the development of therapies. Successful translation of basic research findings into clinical trials and, moreover, approval of the first therapies for blinding inherited and age-related retinal dystrophies has been reported in recent years. This article explores the value of the NHP models in understanding human vision and reviews their contribution to the development of innovative therapeutic strategies to save and restore vision.

5.
Hum Mol Genet ; 28(1): 105-123, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30239717

RESUMEN

Variations in the human Crumbs homolog-1 (CRB1) gene lead to an array of retinal dystrophies including early onset of retinitis pigmentosa (RP) and Leber congenital amaurosis (LCA) in children. To investigate the physiological roles of CRB1 and CRB2 in retinal Müller glial cells (MGCs), we analysed mouse retinas lacking both proteins in MGC. The peripheral retina showed a faster progression of dystrophy than the central retina. The central retina showed retinal folds, disruptions at the outer limiting membrane, protrusion of photoreceptor nuclei into the inner and outer segment layers and ingression of photoreceptor nuclei into the photoreceptor synaptic layer. The peripheral retina showed a complete loss of the photoreceptor synapse layer, intermingling of photoreceptor nuclei within the inner nuclear layer and ectopic photoreceptor cells in the ganglion cell layer. Electroretinography showed severe attenuation of the scotopic a-wave at 1 month of age with responses below detection levels at 3 months of age. The double knockout mouse retinas mimicked a phenotype equivalent to a clinical LCA phenotype due to loss of CRB1. Localization of CRB1 and CRB2 in non-human primate (NHP) retinas was analyzed at the ultrastructural level. We found that NHP CRB1 and CRB2 proteins localized to the subapical region adjacent to adherens junctions at the outer limiting membrane in MGC and photoreceptors. Our data suggest that loss of CRB2 in MGC aggravates the CRB1-associated RP-like phenotype towards an LCA-like phenotype.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Retinitis Pigmentosa/genética , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Modelos Animales de Enfermedad , Electrorretinografía , Células Ependimogliales/metabolismo , Células Ependimogliales/fisiología , Proteínas del Ojo/genética , Proteínas del Ojo/fisiología , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/fisiopatología , Macaca fascicularis , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteínas del Tejido Nervioso/fisiología , Neuroglía/fisiología , Fenotipo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/metabolismo , Distrofias Retinianas/metabolismo , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/fisiopatología
6.
PLoS Comput Biol ; 16(7): e1007857, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32667921

RESUMEN

In many cases of inherited retinal degenerations, ganglion cells are spared despite photoreceptor cell death, making it possible to stimulate them to restore visual function. Several studies have shown that it is possible to express an optogenetic protein in ganglion cells and make them light sensitive, a promising strategy to restore vision. However the spatial resolution of optogenetically-reactivated retinas has rarely been measured, especially in the primate. Since the optogenetic protein is also expressed in axons, it is unclear if these neurons will only be sensitive to the stimulation of a small region covering their somas and dendrites, or if they will also respond to any stimulation overlapping with their axon, dramatically impairing spatial resolution. Here we recorded responses of mouse and macaque retinas to random checkerboard patterns following an in vivo optogenetic therapy. We show that optogenetically activated ganglion cells are each sensitive to a small region of visual space. A simple model based on this small receptive field predicted accurately their responses to complex stimuli. From this model, we simulated how the entire population of light sensitive ganglion cells would respond to letters of different sizes. We then estimated the maximal acuity expected by a patient, assuming it could make an optimal use of the information delivered by this reactivated retina. The obtained acuity is above the limit of legal blindness. Our model also makes interesting predictions on how acuity might vary upon changing the therapeutic strategy, assuming an optimal use of the information present in the retinal activity. Optogenetic therapy could thus potentially lead to high resolution vision, under conditions that our model helps to determinine.


Asunto(s)
Ceguera , Optogenética/métodos , Células Ganglionares de la Retina/fisiología , Animales , Ceguera/fisiopatología , Ceguera/terapia , Terapia Genética , Macaca , Ratones , Modelos Biológicos , Retina/fisiología , Agudeza Visual/fisiología
7.
J Neurosci ; 39(18): 3484-3497, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30833505

RESUMEN

To better examine circuit mechanisms underlying perception and behavior, researchers need tools to enable temporally precise control of action-potential generation of individual cells from neuronal ensembles. Here we demonstrate that such precision can be achieved with two-photon (2P) temporally focused computer-generated holography to control neuronal excitability at the supragranular layers of anesthetized and awake visual cortex in both male and female mice. Using 2P-guided whole-cell or cell-attached recordings in positive neurons expressing any of the three opsins ReaChR, CoChR, or ChrimsonR, we investigated the dependence of spiking activity on the opsin's channel kinetics. We found that in all cases the use of brief illumination (≤10 ms) induces spikes of millisecond temporal resolution and submillisecond precision, which were preserved upon repetitive illuminations up to tens of hertz. To reach high temporal precision, we used a large illumination spot covering the entire cell body and an amplified laser at high peak power and low excitation intensity (on average ≤0.2 mW/µm2), thus minimizing the risk for nonlinear photodamage effects. Finally, by combining 2P holographic excitation with electrophysiological recordings and calcium imaging using GCaMP6s, we investigated the factors, including illumination shape and intensity, opsin distribution in the target cell, and cell morphology, which affect the spatial selectivity of single-cell and multicell holographic activation. Parallel optical control of neuronal activity with cellular resolution and millisecond temporal precision should make it easier to investigate neuronal connections and find further links between connectivity, microcircuit dynamics, and brain functions.SIGNIFICANCE STATEMENT Recent developments in the field of optogenetics has enabled researchers to probe the neuronal microcircuit with light by optically actuating genetically encoded light-sensitive opsins expressed in the target cells. Here, we applied holographic light shaping and temporal focusing to simultaneously deliver axially confined holographic patterns to opsin-positive cells in the living mouse cortex. Parallel illumination efficiently induced action potentials with high temporal resolution and precision for three opsins of different kinetics. We extended the parallel optogenetic activation at low intensity to multiple neurons and concurrently monitored their calcium dynamics. These results demonstrate fast and temporally precise in vivo control of a neuronal subpopulation, opening new opportunities for revealing circuit mechanisms underlying brain functions.


Asunto(s)
Potenciales de Acción , Neuronas/fisiología , Optogenética/métodos , Corteza Visual/fisiología , Animales , Femenino , Luz , Masculino , Ratones Transgénicos , Optogenética/instrumentación , Factores de Tiempo
8.
Biochem Biophys Res Commun ; 527(2): 325-330, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31982136

RESUMEN

Optogenetics is a biological technique that combines the advantageous spatial-temporal resolution of optics and genetic cell targeting to control cellular activity with unprecedented precision. It has found vast applications both in neurosciences and therapy, particularly in view of its application to restore vision in blind patients. Optogenetics requires the ectopic expression of a so-called opsin to render neurons sensitive to light. There are two types of opsins for modulating membrane potential of neurons: (i) microbial opsins from unicellular organisms that respond to a light stimulus by mediating a flow of ions across the membrane (ii) animal opsins that are naturally present in mammalian retinas that initiate G protein coupled signaling in response to light. The former category has been extensively employed for vision restoration in the past decade with two ongoing clinical trials employing microbial opsins to restore light sensation in retinitis pigmentosa patients. The latter subtype of animal opsins is emerging more recently as strong candidates to restore vision with the promise of greater light sensitivity and tolerability. In this review we will discuss each approach in view of its utility for vision restoration in retinal blindness.


Asunto(s)
Ceguera/terapia , Opsinas/genética , Optogenética/métodos , Degeneración Retiniana/terapia , Animales , Ceguera/genética , Ceguera/fisiopatología , Humanos , Retina/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/fisiopatología , Visión Ocular
9.
Int J Mol Sci ; 21(3)2020 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-32028585

RESUMEN

Human induced pluripotent stem cells (hiPSCs) promise a great number of future applications to investigate retinal development, pathophysiology and cell therapies for retinal degenerative diseases. Specific approaches to genetically modulate hiPSC would be valuable for all of these applications. Vectors based on adeno-associated virus (AAV) have shown the ability for gene delivery to retinal organoids derived from hiPSCs. Thus far, little work has been carried out to investigate mechanisms of AAV-mediated gene delivery and the potential advantages of engineered AAVs to genetically modify retinal organoids. In this study, we compared the early transduction efficiency of several recombinant and engineered AAVs in hiPSC-derived RPE cells and retinal organoids in relation to the availability of their cell-surface receptors and as a function of time. The genetic variant AAV2-7m8 had a superior transduction efficiency when applied at day 44 of differentiation on retinal organoids and provided long-lasting expressions for at least 4 weeks after infection without compromising cell viability. All of the capsids we tested transduced the hiPSC-RPE cells, with the AAV2-7m8 variant being the most efficient. Transduction efficiency was correlated with the presence of primary cell-surface receptors on the hiPS-derived organoids. Our study explores some of the mechanisms of cell attachment of AAVs and reports long-term gene expression resulting from gene delivery in retinal organoids.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Pluripotentes Inducidas/citología , Organoides/metabolismo , Retina/metabolismo , Animales , Terapia Genética , Variación Genética , Proteínas Fluorescentes Verdes/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Organoides/citología , Receptores de Superficie Celular/metabolismo , Retina/citología , Transducción Genética , Transgenes
11.
Hum Mol Genet ; 25(14): 3070-3079, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27288449

RESUMEN

Dystrophin-Dp71 being a key membrane cytoskeletal protein, expressed mainly in Müller cells that provide a mechanical link at the Müller cell membrane by direct binding to actin and a transmembrane protein complex. Its absence has been related to blood-retinal barrier (BRB) permeability through delocalization and down-regulation of the AQP4 and Kir4.1 channels (1). We have previously shown that the adeno-associated virus (AAV) variant, ShH10, transduces Müller cells in the Dp71-null mouse retina efficiently and specifically (2,3). Here, we use ShH10 to restore Dp71 expression in Müller cells of Dp71 deficient mouse to study molecular and functional effects of this restoration in an adult mouse displaying retinal permeability. We show that strong and specific expression of exogenous Dp71 in Müller cells leads to correct localization of Dp71 protein restoring all protein interactions in order to re-establish a proper functional BRB and retina homeostasis thus preventing retina from oedema. This study is the basis for the development of new therapeutic strategies in dealing with diseases with BRB breakdown and macular oedema such as diabetic retinopathy (DR).


Asunto(s)
Barrera Hematorretinal/efectos de los fármacos , Distrofina/genética , Edema/terapia , Terapia Genética , Animales , Dependovirus/genética , Distrofina/deficiencia , Distrofina/uso terapéutico , Edema/genética , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/metabolismo , Humanos , Ratones , Ratones Noqueados , Retina/crecimiento & desarrollo , Retina/patología
12.
Mol Ther ; 25(11): 2546-2560, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28807567

RESUMEN

The majority of inherited retinal degenerations converge on the phenotype of photoreceptor cell death. Second- and third-order neurons are spared in these diseases, making it possible to restore retinal light responses using optogenetics. Viral expression of channelrhodopsin in the third-order neurons under ubiquitous promoters was previously shown to restore visual function, albeit at light intensities above illumination safety thresholds. Here, we report (to our knowledge, for the first time) activation of macaque retinas, up to 6 months post-injection, using channelrhodopsin-Ca2+-permeable channelrhodopsin (CatCh) at safe light intensities. High-level CatCh expression was achieved due to a new promoter based on the regulatory region of the gamma-synuclein gene (SNCG) allowing strong expression in ganglion cells across species. Our promoter, in combination with clinically proven adeno-associated virus 2 (AAV2), provides CatCh expression in peri-foveolar ganglion cells responding robustly to light under the illumination safety thresholds for the human eye. On the contrary, the threshold of activation and the proportion of unresponsive cells were much higher when a ubiquitous promoter (cytomegalovirus [CMV]) was used to express CatCh. The results of our study suggest that the inclusion of optimized promoters is key in the path to clinical translation of optogenetics.


Asunto(s)
Channelrhodopsins/genética , Vectores Genéticos/administración & dosificación , Regiones Promotoras Genéticas , Recuperación de la Función , Degeneración Retiniana/terapia , Animales , Channelrhodopsins/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Inyecciones Intravítreas , Luz , Macaca fascicularis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Optogenética , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Transducción Genética , Transgenes , Visión Ocular/fisiología
13.
Neural Plast ; 2017: 6818970, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29234527

RESUMEN

The lack of axonal regeneration and neuronal cell death causes permanent neurological deficits in the injured CNS. Using the classical CNS injury model of optic nerve crush in mice, ciliary neurotrophic factor (CNTF) was found to stimulate retinal ganglion cell (RGC) survival and axonal growth, but in an incomplete fashion. The elucidation of molecular mechanisms impairing CNTF-induced axonal regeneration is paramount to promote visual recovery. In the present study, we sought to evaluate the contribution of sphingosine 1-phosphate receptor 1 (S1PR1) to the neuroprotective and regenerative effects of CNTF. The transduction of retinal cells with adeno-associated viruses (AAV) allowed to activate CNTF/signal transducer and activator of transcription 3 (Stat3) signaling and to modulate S1PR1 expression in RGCs. Our results showed that CNTF/Stat3 prevented injury-induced S1PR1 downregulation. Silencing S1PR1 in RGCs significantly enhanced CNTF-induced axonal growth in the injured optic nerve. In contrast, RGC survival was markedly decreased when S1PR1 was repressed with viral vectors. The level of phosphorylated Stat3 (P-Stat3), an intracellular mediator of CNTF, did not fluctuate after S1PR1 inhibition and CNTF stimulation. Collectively, these results suggest that S1PR1 acts as a major regulator of retinal neuron survival and restricts the RGC growth response induced by CNTF.


Asunto(s)
Axones/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Factor Neurotrófico Ciliar/farmacología , Regeneración Nerviosa/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Receptores de Lisoesfingolípidos/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Axones/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Ratones , Regeneración Nerviosa/fisiología , Neuroprotección/efectos de los fármacos , Fosforilación/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
14.
J Neurosci ; 35(23): 8882-95, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26063920

RESUMEN

Spinocerebellar ataxia type 6 (SCA6) is linked to poly-glutamine (polyQ) within the C terminus (CT) of the pore-forming subunits of P/Q-type Ca(2+) channels (Cav2.1) and is characterized by CT protein aggregates found in cerebellar Purkinje cells (PCs). One hypothesis regarding SCA6 disease is that a CT fragment of the Cav2.1 channel, which is detected specifically in cytosolic and nuclear fractions in SCA6 patients, is associated with the SCA6 pathogenesis. To test this hypothesis, we expressed P/Q-type channel protein fragments from two different human CT splice variants, as predicted from SCA6 patients, in PCs of mice using viral and transgenic approaches. These splice variants represent a short (CT-short without polyQs) and a long (CT-long with 27 polyQs) CT fragment. Our results show that the different splice variants of the CTs differentially distribute within PCs, i.e., the short CTs reveal predominantly nuclear inclusions, whereas the long CTs prominently reveal both nuclear and cytoplasmic aggregates. Postnatal expression of CTs in PCs in mice reveals that only CT-long causes SCA6-like symptoms, i.e., deficits in eyeblink conditioning (EBC), ataxia, and PC degeneration. The physiological phenotypes associated specifically with the long CT fragment can be explained by an impairment of LTD and LTP at the parallel fiber-to-PC synapse and alteration in spontaneous PC activity. Thus, our results suggest that the polyQ carrying the CT fragment of the P/Q-type channel is sufficient to cause SCA6 pathogenesis in mice and identifies EBC as a new diagnostic strategy to evaluate Ca(2+) channel-mediated human diseases.


Asunto(s)
Cerebelo/patología , Discapacidades para el Aprendizaje/genética , Plasticidad Neuronal/genética , Neuronas/fisiología , Agregado de Proteínas/genética , Ataxias Espinocerebelosas/metabolismo , Actinas/metabolismo , Potenciales de Acción/genética , Factores de Edad , Animales , Proteínas Bacterianas/genética , Canales de Calcio Tipo N/genética , Corteza Cerebelosa/patología , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Células HEK293 , Humanos , Técnicas In Vitro , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Transgénicos , Actividad Motora/genética , Neuronas/patología , Péptidos/genética , Desempeño Psicomotor/fisiología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología
15.
Biotechnol Bioeng ; 113(12): 2712-2724, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27259396

RESUMEN

Recently, we described a modified AAV2 vector-AAV2-7m8-having a capsid-displayed peptide insertion of 10 amino acids with enhanced retinal transduction properties. The insertion of the peptide referred to as 7m8 is responsible for high-level gene delivery into deep layers of the retina when virus is delivered into the eye's vitreous. Here, we further characterize AAV2-7m8 mediated gene delivery to neural tissue and investigate the mechanisms by which the inserted peptide provides better transduction away from the injection site. First, in order to understand if the peptide exerts its effect on its own or in conjunction with the neighboring amino acids, we inserted the 7m8 peptide at equivalent positions on three other AAV capsids, AAV5, AAV8, and AAV9, and evaluated its effect on their infectivity. Intravitreal delivery of these peptide insertion vectors revealed that only AAV9 benefited from 7m8 insertion in the context of the retina. We then investigated AAV2-7m8 and AAV9-7m8 properties in the brain, to better evaluate the spread and efficacy of viral transduction in view of the peptide insertion. While 7m8 insertion led to higher intensity gene expression, the spread of gene expression remained unchanged compared to the parental serotypes. Our results indicate that the 7m8 peptide insertion acts by increasing efficacy of cellular entry, with little effect on the spread of viral particles in neural tissue. The effects of peptide insertion are capsid and tissue dependent, highlighting the importance of the microenvironment in gene delivery using AAV. Biotechnol. Bioeng. 2016;113: 2712-2724. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteínas de la Cápside/genética , Dependovirus/genética , Vectores Genéticos/genética , Proteínas Recombinantes/metabolismo , Retina/virología , Transducción Genética/métodos , Animales , Proteínas de la Cápside/química , Proteínas de la Cápside/ultraestructura , Dependovirus/química , Dependovirus/ultraestructura , Variación Genética/genética , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Conformación Molecular , Proteínas Recombinantes/genética , Retina/fisiología
16.
Mol Ther ; 23(1): 7-16, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25095892

RESUMEN

Most inherited retinal dystrophies display progressive photoreceptor cell degeneration leading to severe visual impairment. Optogenetic reactivation of retinal neurons mediated by adeno-associated virus (AAV) gene therapy has the potential to restore vision regardless of patient-specific mutations. The challenge for clinical translatability is to restore a vision as close to natural vision as possible, while using a surgically safe delivery route for the fragile degenerated retina. To preserve the visual processing of the inner retina, we targeted ON bipolar cells, which are still present at late stages of disease. For safe gene delivery, we used a recently engineered AAV variant that can transduce the bipolar cells after injection into the eye's easily accessible vitreous humor. We show that AAV encoding channelrhodopsin under the ON bipolar cell-specific promoter mediates long-term gene delivery restricted to ON-bipolar cells after intravitreal administration. Channelrhodopsin expression in ON bipolar cells leads to restoration of ON and OFF responses at the retinal and cortical levels. Moreover, light-induced locomotory behavior is restored in treated blind mice. Our results support the clinical relevance of a minimally invasive AAV-mediated optogenetic therapy for visual restoration.


Asunto(s)
Ceguera/terapia , Dependovirus/genética , Terapia Genética/métodos , Células Bipolares de la Retina/metabolismo , Degeneración Retiniana/terapia , Animales , Conducta Animal , Ceguera/genética , Ceguera/patología , Channelrhodopsins , Femenino , Expresión Génica , Técnicas de Transferencia de Gen , Ingeniería Genética , Vectores Genéticos , Inyecciones Intravítreas , Luz , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas , Células Bipolares de la Retina/patología , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Percepción Visual/genética , Cuerpo Vítreo
17.
J Neurosci ; 34(19): 6596-605, 2014 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-24806684

RESUMEN

The fovea dominates primate vision, and its anatomy and perceptual abilities are well studied, but its physiology has been little explored because of limitations of current physiological methods. In this study, we adapted a novel in vivo imaging method, originally developed in mouse retina, to explore foveal physiology in the macaque, which permits the repeated imaging of the functional response of many retinal ganglion cells (RGCs) simultaneously. A genetically encoded calcium indicator, G-CaMP5, was inserted into foveal RGCs, followed by calcium imaging of the displacement of foveal RGCs from their receptive fields, and their intensity-response functions. The spatial offset of foveal RGCs from their cone inputs makes this method especially appropriate for fovea by permitting imaging of RGC responses without excessive light adaptation of cones. This new method will permit the tracking of visual development, progression of retinal disease, or therapeutic interventions, such as insertion of visual prostheses.


Asunto(s)
Ojo/efectos de la radiación , Fóvea Central/efectos de la radiación , Luz , Células Ganglionares de la Retina/efectos de la radiación , Anestesia , Animales , Dependovirus/genética , Fenómenos Electrofisiológicos , Ojo/anatomía & histología , Movimientos Oculares/fisiología , Femenino , Colorantes Fluorescentes , Fóvea Central/citología , Técnicas de Transferencia de Gen , Fototransducción/fisiología , Macaca fascicularis , Microscopía Confocal , Neuroimagen/métodos , Estimulación Luminosa , Células Fotorreceptoras de Vertebrados/fisiología , Retina/crecimiento & desarrollo , Degeneración Retiniana/patología , Relación Señal-Ruido
18.
Curr Opin Neurol ; 28(1): 51-60, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25545056

RESUMEN

PURPOSE OF REVIEW: This review will discuss retinal gene therapy strategies with a focus on mutation-independent approaches to treat a large number of patients without knowledge of the mutant gene. These approaches rely on the secretion of neurotrophic factors to slow down retinal degeneration and the use of optogenetics to restore vision in late-stage disease. RECENT FINDINGS: Success in clinical application of adeno-associated virus (AAV)-mediated gene therapy for Leber's congenital amaurosis established the feasibility of retinal gene therapy. More clinical trials are currently on their way for recessive diseases with known mutations. However, the genetic and mechanistic diversity of the retinal diseases presents an enormous obstacle for the development of gene therapies tailored to each patient-specific mutation. To extend gene therapy's promise to a large number of patients, evidence suggests retina-specific trophic factors, such as rod-derived cone viability factor, can be used to slow down loss of cone cells responsible for our high acuity vision. In parallel, it has been shown that microbial opsins are able to restore light sensitivity when expressed in blind retinas. SUMMARY: Recent findings imply that using the viral technology that has been demonstrated as well tolerated in patients, there are opportunities to develop widely applicable gene therapeutic interventions in clinical ophthalmology.


Asunto(s)
Proteínas del Ojo/genética , Terapia Genética/métodos , Mutación , Enfermedades de la Retina/terapia , Dependovirus , Vectores Genéticos , Humanos , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/terapia , Degeneración Retiniana/genética , Degeneración Retiniana/terapia , Enfermedades de la Retina/genética
19.
Nat Methods ; 9(4): 396-402, 2012 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-22343342

RESUMEN

Local anesthetics effectively suppress pain sensation, but most of these compounds act nonselectively, inhibiting activity of all neurons. Moreover, their actions abate slowly, preventing precise spatial and temporal control of nociception. We developed a photoisomerizable molecule, quaternary ammonium-azobenzene-quaternary ammonium (QAQ), that enables rapid and selective optical control of nociception. QAQ is membrane-impermeant and has no effect on most cells, but it infiltrates pain-sensing neurons through endogenous ion channels that are activated by noxious stimuli, primarily TRPV1. After QAQ accumulates intracellularly, it blocks voltage-gated ion channels in the trans form but not the cis form. QAQ enables reversible optical silencing of mouse nociceptive neuron firing without exogenous gene expression and can serve as a light-sensitive analgesic in rats in vivo. Because intracellular QAQ accumulation is a consequence of nociceptive ion-channel activity, QAQ-mediated photosensitization is a platform for understanding signaling mechanisms in acute and chronic pain.


Asunto(s)
Canales Iónicos/metabolismo , Nocicepción/efectos de los fármacos , Nocicepción/efectos de la radiación , Animales , Compuestos Azo/química , Compuestos Azo/farmacología , Línea Celular , Células HEK293 , Humanos , Canales Iónicos/antagonistas & inhibidores , Ratones , Terminaciones Nerviosas/efectos de los fármacos , Terminaciones Nerviosas/efectos de la radiación , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de la radiación , Estimulación Luminosa , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Compuestos de Amonio Cuaternario/química , Compuestos de Amonio Cuaternario/farmacología , Ratas , Receptores Purinérgicos P2X7/metabolismo , Médula Espinal/citología , Médula Espinal/efectos de los fármacos , Médula Espinal/efectos de la radiación , Canales Catiónicos TRPV/metabolismo , Factores de Tiempo
20.
Med Sci (Paris) ; 31(5): 529-37, 2015 May.
Artículo en Francés | MEDLINE | ID: mdl-26059304

RESUMEN

Gene therapy is quickly becoming a reality applicable in the clinic for inherited retinal diseases. Its remarkable success in safety and efficacy, in clinical trials for Leber's congenital amaurosis (LCA) type II generated significant interest and opened up possibilities for a new era of retinal gene therapies. Success in these clinical trials was mainly due to the favorable characteristics of the retina as a target organ. The eye offers several advantages as it is readily accessible and has some degree of immune privilege making it suitable for application of viral vectors. The viral vectors most frequently used for retinal gene delivery are lentivirus, adenovirus and adeno-associated virus (AAV). Here we will discuss the use of these viral vectors in retinal gene delivery with a strong focus on favorable properties of AAV. Thanks to its small size, AAV diffuses well in the inter-neural matrix making it suitable for applications in neural retina. Building on this initial clinical success with LCA II, we have now many opportunities to extend this proof-of-concept to other retinal diseases using AAV as a vector. This article will discuss what are some of the most imminent cellular targets for such therapies and the AAV toolkit that has been built to target these cells successfully. We will also discuss some of the challenges that we face in translating AAV-based gene therapies to the clinic.


Asunto(s)
Oftalmopatías/terapia , Terapia Genética , Vectores Genéticos/uso terapéutico , Adenoviridae/genética , Adenoviridae/fisiología , Animales , Cápside , ADN Viral/genética , Dependovirus/genética , Dependovirus/fisiología , Evolución Molecular Dirigida , Oftalmopatías/genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Inyecciones Intraoculares , Lentivirus/genética , Lentivirus/fisiología , Ratones , Mutagénesis Insercional , Mutagénesis Sitio-Dirigida , Primates , ARN Viral/genética , Receptores Virales/fisiología , Retina/ultraestructura , Transducción Genética , Transgenes , Investigación Biomédica Traslacional , Tropismo Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA