Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(13): e2117038119, 2022 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-35316139

RESUMO

SignificanceCanine models of inherited retinal diseases have helped advance adeno-associated virus (AAV)-based gene therapies targeting specific cells in the outer retina for treating blinding diseases in patients. However, therapeutic targeting of diseases such as congenital stationary night blindness (CSNB) that exhibit defects in ON-bipolar cells (ON-BCs) of the midretina remains underdeveloped. Using a leucine-rich repeat, immunoglobulin-like and transmembrane domain 3 (LRIT3) mutant canine model of CSNB exhibiting ON-BC dysfunction, we tested the ability of cell-specific AAV capsids and promotors to specifically target ON-BCs for gene delivery. Subretinal injection of one vector demonstrated safety and efficacy with robust and stable rescue of electroretinography signals and night vision up to 1 y, paving the way for clinical trials in patients.


Assuntos
Doenças Genéticas Ligadas ao Cromossomo X , Cegueira Noturna , Animais , Dependovirus/genética , Cães , Eletrorretinografia , Oftalmopatias Hereditárias , Doenças Genéticas Ligadas ao Cromossomo X/genética , Doenças Genéticas Ligadas ao Cromossomo X/terapia , Terapia Genética , Humanos , Proteínas de Membrana/genética , Miopia , Cegueira Noturna/genética , Cegueira Noturna/terapia
2.
Nat Methods ; 16(10): 1063, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31501552

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Nat Methods ; 16(9): 853-857, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31427757

RESUMO

Single-molecule localization microscopy (SMLM), while well established for cultured cells, is not yet fully compatible with tissue-scale samples. We introduce single-molecule oblique-plane microscopy (obSTORM), which by directly imaging oblique sections of samples with oblique light-sheet illumination offers a deep and volumetric SMLM platform that is convenient for standard tissue samples and small intact animals. We demonstrate super-resolution imaging at depths of up to 66 µm for cells, Caenorhabditis elegans gonads, Drosophila melanogaster larval brain, mouse retina and brain sections, and whole stickleback fish.


Assuntos
Encéfalo/diagnóstico por imagem , Caenorhabditis elegans/metabolismo , Drosophila melanogaster/metabolismo , Peixes/metabolismo , Microscopia de Fluorescência/métodos , Retina/diagnóstico por imagem , Imagem Individual de Molécula/métodos , Células A549 , Animais , Feminino , Humanos , Imageamento Tridimensional , Masculino , Camundongos , Camundongos Endogâmicos C57BL
4.
Ophthalmic Res ; 2022 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-36103843

RESUMO

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.

5.
J Neurosci ; 38(9): 2246-2261, 2018 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-29378863

RESUMO

Müller glial (MG) cells in the zebrafish retina respond to injury by acquiring retinal stem-cell characteristics. Thousands of gene expression changes are associated with this event. Key among these changes is the induction of Ascl1a and Lin28a, two reprogramming factors whose expression is necessary for retina regeneration. Whether these factors are sufficient to drive MG proliferation and subsequent neuronal-fate specification remains unknown. To test this, we conditionally expressed Ascl1a and Lin28a in the uninjured retina of male and female fish. We found that together, their forced expression only stimulates sparse MG proliferation. However, in combination with Notch signaling inhibition, widespread MG proliferation and neuron regeneration ensued. Remarkably, Ascl1 and Lin28a expression in the retina of male and female mice also stimulated sparse MG proliferation, although this was not enhanced when combined with inhibitors of Notch signaling. Lineage tracing in both fish and mice suggested that the proliferating MG generated multipotent progenitors; however, this process was much more efficient in fish than mice. Overall, our studies suggest that the overexpression of Ascl1a and Lin28a in zebrafish, in combination with inhibition of Notch signaling, can phenocopy the effects of retinal injury in Müller glia. Interestingly, Ascl1 and Lin28a seem to have similar effects in fish and mice, whereas Notch signaling may differ. Understanding the different consequences of Notch signaling inhibition in fish and mice, may suggest additional strategies for enhancing retina regeneration in mammals.SIGNIFICANCE STATEMENT Mechanisms underlying retina regeneration in fish may suggest strategies for stimulating this process in mammals. Here we report that forced expression of Ascl1 and Lin28a can stimulate sparse MG proliferation in fish and mice; however, only in fish does Notch signaling inhibition collaborate with Ascl1a and Lin28a to stimulate widespread MG proliferation in the uninjured retina. Discerning differences in Notch signaling between fish and mice MG may reveal strategies for stimulating retina regeneration in mammals.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regeneração Nervosa/fisiologia , Proteínas de Ligação a RNA/metabolismo , Receptores Notch/metabolismo , Retina/fisiologia , Animais , Proliferação de Células/fisiologia , Células Ependimogliais/metabolismo , Feminino , Masculino , Camundongos , Neurogênese/fisiologia , Peixe-Zebra
6.
Genome Res ; 26(2): 238-55, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26576614

RESUMO

Cis-regulatory elements (CREs, e.g., promoters and enhancers) regulate gene expression, and variants within CREs can modulate disease risk. Next-generation sequencing has enabled the rapid generation of genomic data that predict the locations of CREs, but a bottleneck lies in functionally interpreting these data. To address this issue, massively parallel reporter assays (MPRAs) have emerged, in which barcoded reporter libraries are introduced into cells, and the resulting barcoded transcripts are quantified by next-generation sequencing. Thus far, MPRAs have been largely restricted to assaying short CREs in a limited repertoire of cultured cell types. Here, we present two advances that extend the biological relevance and applicability of MPRAs. First, we adapt exome capture technology to instead capture candidate CREs, thereby tiling across the targeted regions and markedly increasing the length of CREs that can be readily assayed. Second, we package the library into adeno-associated virus (AAV), thereby allowing delivery to target organs in vivo. As a proof of concept, we introduce a capture library of about 46,000 constructs, corresponding to roughly 3500 DNase I hypersensitive (DHS) sites, into the mouse retina by ex vivo plasmid electroporation and into the mouse cerebral cortex by in vivo AAV injection. We demonstrate tissue-specific cis-regulatory activity of DHSs and provide examples of high-resolution truncation mutation analysis for multiplex parsing of CREs. Our approach should enable massively parallel functional analysis of a wide range of CREs in any organ or species that can be infected by AAV, such as nonhuman primates and human stem cell-derived organoids.


Assuntos
Córtex Cerebral/metabolismo , Regiões Promotoras Genéticas , Animais , Sequência de Bases , Análise Mutacional de DNA , Dependovirus/genética , Epigênese Genética , Feminino , Biblioteca Gênica , Loci Gênicos , Vetores Genéticos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Retina/metabolismo , Transdução Genética
7.
Exp Eye Res ; 167: 56-90, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29122605

RESUMO

We produced 8 lines of transgenic (Tg) rats expressing one of two different rhodopsin mutations in albino Sprague-Dawley (SD) rats. Three lines were generated with a proline to histidine substitution at codon 23 (P23H), the most common autosomal dominant form of retinitis pigmentosa in the United States. Five lines were generated with a termination codon at position 334 (S334ter), resulting in a C-terminal truncated opsin protein lacking the last 15 amino acid residues and containing all of the phosphorylation sites involved in rhodopsin deactivation, as well as the terminal QVAPA residues important for rhodopsin deactivation and trafficking. The rates of photoreceptor (PR) degeneration in these models vary in proportion to the ratio of mutant to wild-type rhodopsin. The models have been widely studied, but many aspects of their phenotypes have not been described. Here we present a comprehensive study of the 8 Tg lines, including the time course of PR degeneration from the onset to one year of age, retinal structure by light and electron microscopy (EM), hemispheric asymmetry and gradients of rod and cone degeneration, rhodopsin content, gene dosage effect, rapid activation and invasion of the outer retina by presumptive microglia, rod outer segment disc shedding and phagocytosis by the retinal pigmented epithelium (RPE), and retinal function by the electroretinogram (ERG). The biphasic nature of PR cell death was noted, as was the lack of an injury-induced protective response in the rat models. EM analysis revealed the accumulation of submicron vesicular structures in the interphotoreceptor space during the peak period of PR outer segment degeneration in the S334ter lines. This is likely due to the elimination of the trafficking consensus domain as seen before as with other rhodopsin mutants lacking the C-terminal QVAPA. The 8 rhodopsin Tg lines have been, and will continue to be, extremely useful models for the experimental study of inherited retinal degenerations.


Assuntos
Modelos Animais de Doenças , Células Fotorreceptoras de Vertebrados/patologia , Mutação Puntual , Retina/fisiologia , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Rodopsina/genética , Animais , Eletrorretinografia , Microscopia , Microscopia Eletrônica , Fenótipo , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Degeneração Retiniana/fisiopatologia
8.
Nature ; 488(7411): 379-83, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22878719

RESUMO

Inhibitory interneurons are essential components of the neural circuits underlying various brain functions. In the neocortex, a large diversity of GABA (γ-aminobutyric acid) interneurons has been identified on the basis of their morphology, molecular markers, biophysical properties and innervation pattern. However, how the activity of each subtype of interneurons contributes to sensory processing remains unclear. Here we show that optogenetic activation of parvalbumin-positive (PV+) interneurons in the mouse primary visual cortex (V1) sharpens neuronal feature selectivity and improves perceptual discrimination. Using multichannel recording with silicon probes and channelrhodopsin-2 (ChR2)-mediated optical activation, we found that increased spiking of PV+ interneurons markedly sharpened orientation tuning and enhanced direction selectivity of nearby neurons. These effects were caused by the activation of inhibitory neurons rather than a decreased spiking of excitatory neurons, as archaerhodopsin-3 (Arch)-mediated optical silencing of calcium/calmodulin-dependent protein kinase IIα (CAMKIIα)-positive excitatory neurons caused no significant change in V1 stimulus selectivity. Moreover, the improved selectivity specifically required PV+ neuron activation, as activating somatostatin or vasointestinal peptide interneurons had no significant effect. Notably, PV+ neuron activation in awake mice caused a significant improvement in their orientation discrimination, mirroring the sharpened V1 orientation tuning. Together, these results provide the first demonstration that visual coding and perception can be improved by increased spiking of a specific subtype of cortical inhibitory interneurons.


Assuntos
Interneurônios/fisiologia , Córtex Visual/citologia , Córtex Visual/fisiologia , Percepção Visual/fisiologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/deficiência , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Channelrhodopsins , Aprendizagem por Discriminação , Camundongos , Modelos Neurológicos , Inibição Neural/fisiologia , Parvalbuminas/metabolismo , Rodopsinas Microbianas/metabolismo , Vigília/fisiologia , Ácido gama-Aminobutírico/metabolismo
9.
Adv Exp Med Biol ; 1074: 75-81, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29721930

RESUMO

The clinical success of gene replacement therapies in recent years has served as a proof of concept for the treatment of inherited retinal degenerations using adeno-associated virus (AAV) as viral vector. However, inherited retinal degenerative diseases showcase a broad genetic and mechanistic heterogeneity, challenging the development of mutation-specific therapies for each specific mutation. Mutation-independent approaches must be developed to slow down retinal degeneration regardless of the underlying genetic mutation and onset of the disease. New understanding of cell death mechanisms in rod-cone dystrophies have led to promising rescue of photoreceptor cell death by virally mediating expression of anti-apoptotic factors and secretion of retinal neurotrophic factors. Optogenetic therapies are also able to restore light sensitivities in blind retinas.


Assuntos
Distrofias de Cones e Bastonetes/terapia , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Optogenética/métodos , Morte Celular , Channelrhodopsins/genética , Channelrhodopsins/uso terapêutico , Distrofias de Cones e Bastonetes/genética , Dependovirus/genética , Progressão da Doença , Células Ependimogliais/metabolismo , Humanos , Mutação , Fatores de Crescimento Neural/metabolismo , Células Fotorreceptoras Retinianas Cones/patologia , Células Fotorreceptoras Retinianas Bastonetes/patologia , Transdução Genética
10.
Hum Mol Genet ; 24(11): 3104-18, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25701872

RESUMO

Mutations in the Crumbs-homologue-1 (CRB1) gene lead to severe recessive inherited retinal dystrophies. Gene transfer therapy is the most promising cure for retinal dystrophies and has primarily been applied for recessive null conditions via a viral gene expression vector transferring a cDNA encoding an enzyme or channel protein, and targeting expression to one cell type. Therapy for the human CRB1 disease will be more complex, as CRB1 is a structural and signaling transmembrane protein present in three cell classes: Müller glia, cone and rod photoreceptors. In this study, we applied CRB1 and CRB2 gene therapy vectors in Crb1-retinitis pigmentosa mouse models at mid-stage disease. We tested if CRB expression restricted to Müller glial cells or photoreceptors or co-expression in both is required to recover retinal function. We show that targeting both Müller glial cells and photoreceptors with CRB2 ameliorated retinal function and structure in Crb1 mouse models. Surprisingly, targeting a single cell type or all cell types with CRB1 reduced retinal function. We show here the first pre-clinical studies for CRB1-related eye disorders using CRB2 vectors and initial elucidation of the cellular mechanisms underlying CRB1 function.


Assuntos
Células Ependimogliais/fisiologia , Proteínas do Tecido Nervoso/genética , Retinose Pigmentar/genética , Animais , Proteínas de Transporte/genética , Modelos Animais de Doenças , Terapia Genética , Células HEK293 , Humanos , Injeções Intravítreas , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Retina/patologia , Retina/fisiopatologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Retinose Pigmentar/terapia
11.
Proc Natl Acad Sci U S A ; 111(51): E5574-83, 2014 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-25489083

RESUMO

Most inherited forms of blindness are caused by mutations that lead to photoreceptor cell death but spare second- and third-order retinal neurons. Expression of the light-gated excitatory mammalian ion channel light-gated ionotropic glutamate receptor (LiGluR) in retinal ganglion cells (RGCs) of the retina degeneration (rd1) mouse model of blindness was previously shown to restore some visual functions when stimulated by UV light. Here, we report restored retinal function in visible light in rodent and canine models of blindness through the use of a second-generation photoswitch for LiGluR, maleimide-azobenzene-glutamate 0 with peak efficiency at 460 nm (MAG0(460)). In the blind rd1 mouse, multielectrode array recordings of retinal explants revealed robust and uniform light-evoked firing when LiGluR-MAG0(460) was targeted to RGCs and robust but diverse activity patterns in RGCs when LiGluR-MAG0(460) was targeted to ON-bipolar cells (ON-BCs). LiGluR-MAG0(460) in either RGCs or ON-BCs of the rd1 mouse reinstated innate light-avoidance behavior and enabled mice to distinguish between different temporal patterns of light in an associative learning task. In the rod-cone dystrophy dog model of blindness, LiGluR-MAG0(460) in RGCs restored robust light responses to retinal explants and intravitreal delivery of LiGluR and MAG0(460) was well tolerated in vivo. The results in both large and small animal models of photoreceptor degeneration provide a path to clinical translation.


Assuntos
Ativação do Canal Iônico , Canais Iônicos/efeitos da radiação , Luz , Células Ganglionares da Retina/efeitos da radiação , Visão Ocular , Animais , Cegueira/fisiopatologia , Canais Iônicos/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Células Ganglionares da Retina/fisiologia
12.
Hum Mol Genet ; 23(13): 3384-401, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24493795

RESUMO

In humans, the Crumbs homolog-1 (CRB1) gene is mutated in autosomal recessive Leber congenital amaurosis and early-onset retinitis pigmentosa. In mammals, the Crumbs family is composed of: CRB1, CRB2, CRB3A and CRB3B. Recently, we showed that removal of mouse Crb2 from retinal progenitor cells, and consequent removal from Müller glial and photoreceptor cells, results in severe and progressive retinal degeneration with concomitant loss of retinal function that mimics retinitis pigmentosa due to mutations in the CRB1 gene. Here, we studied the effects of cell-type-specific loss of CRB2 from the developing mouse retina using targeted conditional deletion of Crb2 in photoreceptors or Müller cells. We analyzed the consequences of targeted loss of CRB2 in the adult mouse retina using adeno-associated viral vectors encoding Cre recombinase and short hairpin RNA against Crb2. In vivo retinal imaging by means of optical coherence tomography on retinas lacking CRB2 in photoreceptors showed progressive thinning of the photoreceptor layer and cellular mislocalization. Electroretinogram recordings under scotopic conditions showed severe attenuation of the a-wave, confirming the degeneration of photoreceptors. Retinas lacking CRB2 in developing photoreceptors showed early onset of abnormal lamination, whereas retinas lacking CRB2 in developing Müller cells showed late onset retinal disorganization. Our data suggest that in the developing retina, CRB2 has redundant functions in Müller glial cells, while CRB2 has essential functions in photoreceptors. Our data suggest that short-term loss of CRB2 in adult mouse photoreceptors, but not in Müller glial cells, causes sporadic loss of adhesion between photoreceptors and Müller cells.


Assuntos
Proteínas de Membrana/metabolismo , Células Fotorreceptoras/metabolismo , Retinose Pigmentar/etiologia , Retinose Pigmentar/metabolismo , Animais , Células Ependimogliais/metabolismo , Feminino , Imuno-Histoquímica , Masculino , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Retinose Pigmentar/genética
13.
Mol Ther ; 23(2): 290-6, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25224467

RESUMO

Systemic delivery of AAV9 offers the potential for widespread and efficient gene delivery to the retina, and may thus be a useful approach for treatment of disease where intraocular injections are not possible, for syndromes affecting multiple organs, or where early intervention is required. The expression resulting from intravenous injection of AAV9 is more efficient in neonates than adults, and here we characterize the effect of age on retinal transduction of AAV9 in the mouse retina. We find that the pattern of expression in neonatal mice is correlated to the development of the retinal vasculature, and that the area of the retinal transduction as well as the cell types infected vary depending on the age at injection. Furthermore, we demonstrate that sequential injections of AAV9 vectors carrying two different transgenes infect adjacent areas of the retina, providing a larger area of coverage. Lastly, we show that the retina's endogenous spatiotemporal expression pattern of Mfsd2a, a protein associated with the maturation of a functional blood-brain barrier, coincides with suppression of retinal transduction by intravenously-delivered AAV9, suggesting that AAV9 crosses the blood-retina barrier through transcytosis.


Assuntos
Dependovirus/classificação , Dependovirus/genética , Expressão Gênica , Vetores Genéticos/genética , Retina/metabolismo , Transdução Genética , Fatores Etários , Animais , Encéfalo/metabolismo , Genes Reporter , Vetores Genéticos/administração & dosagem , Injeções Intravenosas , Camundongos , Vasos Retinianos , Transgenes
14.
Mol Ther ; 23(10): 1562-71, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26137852

RESUMO

Retinal disease is one of the most active areas of gene therapy, with clinical trials ongoing in the United States for five diseases. There are currently no treatments for patients with late-stage disease in which photoreceptors have been lost. Optogenetic gene therapies are in development, but, to date, have suffered from the low light sensitivity of microbial opsins, such as channelrhodopsin and halorhodopsin, and azobenzene-based photoswitches. Several groups have shown that photoreceptive G-protein-coupled receptors (GPCRs) can be expressed heterologously, and photoactivate endogenous Gi/o signaling. We hypothesized such a GPCR could increase sensitivity due to endogenous signal amplification. We targeted vertebrate rhodopsin to retinal ON-bipolar cells of blind rd1 mice and observed restoration of: (i) light responses in retinal explants, (ii) visually-evoked potentials in visual cortex in vivo, and (iii) two forms of visually-guided behavior: innate light avoidance and discrimination of temporal light patterns in the context of fear conditioning. Importantly, both the light responses of the retinal explants and the visually-guided behavior occurred reliably at light levels that were two to three orders of magnitude dimmer than required for channelrhodopsin. Thus, gene therapy with native light-gated GPCRs presents a novel approach to impart light sensitivity for visual restoration in a useful range of illumination.


Assuntos
Optogenética/métodos , Rodopsina/genética , Visão Ocular/genética , Animais , Dependovirus/genética , Expressão Ectópica do Gene , Potenciais Evocados Visuais/genética , Potenciais Evocados Visuais/efeitos da radiação , Terapia Genética , Vetores Genéticos/genética , Luz , Camundongos , Estimulação Luminosa , Retina/citologia , Retina/metabolismo , Células Bipolares da Retina/metabolismo , Células Ganglionares da Retina/metabolismo , Transdução Genética , Percepção Visual
15.
J Neurosci ; 34(19): 6596-605, 2014 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24806684

RESUMO

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.


Assuntos
Olho/efeitos da radiação , Fóvea Central/efeitos da radiação , Luz , Células Ganglionares da Retina/efeitos da radiação , Anestesia , Animais , Dependovirus/genética , Fenômenos Eletrofisiológicos , Olho/anatomia & histologia , Movimentos Oculares/fisiologia , Feminino , Corantes Fluorescentes , Fóvea Central/citologia , Técnicas de Transferência de Genes , Transdução de Sinal Luminoso/fisiologia , Macaca fascicularis , Microscopia Confocal , Neuroimagem/métodos , Estimulação Luminosa , Células Fotorreceptoras de Vertebrados/fisiologia , Retina/crescimento & desenvolvimento , Degeneração Retiniana/patologia , Razão Sinal-Ruído
16.
Glia ; 62(3): 468-76, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24382652

RESUMO

Formation and maintenance of the blood-retinal barrier (BRB) is required for proper vision and breaching of this barrier contributes to the pathology in a wide variety of retinal conditions such as retinal detachment and diabetic retinopathy. Dystrophin Dp71 being a key membrane cytoskeletal protein, expressed mainly in Müller cells, its absence has been related to BRB permeability through delocalization and down-regulation of the AQP4 and Kir4.1 channels. Dp71-null mouse is thus an excellent model to approach the study of retinal pathologies showing blood-retinal barrier permeability. We aimed to investigate the participation of Müller cells in the BRB and in the inner limiting membrane of Dp71-null mice compared with wild-type mice in order to understand how these barriers work in this model of permeable BRB. To this aim, we used an Adeno-associated virus (AAV) variant, ShH10-GFP, engineered to target Müller cells specifically. ShH10 coding GFP was introduced by intravitreal injection and Müller cell transduction was studied in Dp71-null mice in comparison to wild-type animals. We show that Müller cell transduction follows a significantly different pattern in Dp71-null mice indicating changes in viral cell-surface receptors as well as differences in the permeability of the inner limiting membrane in this mouse line. However, the compromised BRB of the Dp71-null mice does not lead to virus leakage into the bloodstream when the virus is injected intravitreally - an important consideration for AAV-mediated retinal gene therapy.


Assuntos
Barreira Hematorretiniana/fisiopatologia , Distrofina/deficiência , Células Ependimogliais/metabolismo , Retina/patologia , Doenças Retinianas/patologia , Adenoviridae/genética , Animais , Modelos Animais de Doenças , Distrofina/genética , Fundo de Olho , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Knockout , Células Fotorreceptoras/patologia , Retina/metabolismo , Doenças Retinianas/genética , Tomografia de Coerência Óptica , Vias Visuais/patologia , Vias Visuais/fisiopatologia
17.
Adv Exp Med Biol ; 801: 687-93, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24664759

RESUMO

Adeno-associated virus (AAV) is a small, non-pathogenic dependovirus that has shown great potential for safe and long-term expression of a genetic payload in the retina. AAV has been used to treat a growing number of animal models of inherited retinal degeneration, though drawbacks-including a limited carrying capacity, slow onset of expression, and a limited ability to transduce some retinal cell types from the vitreous-restrict the utility of AAV for treating some forms of inherited eye disease. Next generation AAV vectors are being created to address these needs, through rational design efforts such as the creation of self-complementary AAV vectors for faster onset of expression and specific mutations of surface-exposed residues to increase transduction of viral particles. Furthermore, directed evolution has been used to create, through an iterative process of selection, novel variants of AAV with newly acquired, advantageous characteristics. These novel AAV variants have been shown to improve the therapeutic potential of AAV vectors, and further improvements may be achieved through rational design, directed evolution, or a combination of these approaches, leading to broader applicability of AAV and improved treatments for inherited retinal degeneration.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos , Retina/fisiologia , Doenças Retinianas/terapia , Animais , Terapia Genética/tendências , Humanos
18.
Neurobiol Dis ; 51: 202-13, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23194670

RESUMO

The optic nerve crush injury is a well-accepted model to study the mechanisms of axonal regeneration after trauma in the CNS. The infection of retinal ganglion cells (RGCs) with an adeno-associated virus serotype 2 - ciliary neurotrophic factor (AAV2.CNTF) was previously shown to stimulate axonal regeneration. However, the transfection of axotomized neurons themselves may not be optimal to promote full axonal regeneration in the visual system. Here, we show that the release of CNTF by glial cells is a very powerful stimulus for optic fiber regeneration and RGC survival after optic nerve crush. After 8 weeks, long-distance regeneration of severed optic axons was induced by CNTF until and beyond the optic chiasm. Regenerated axons stayed for at least 6 months in the damaged optic nerve. Strikingly, however, many regenerated axons showed one or several sharp U-turns along their course, suggesting that guidance cues are missing and that long-distance axonal regeneration is limited by the return of the growing axons toward the retina. Even more surprisingly, massive axonal sprouting was observed within the eye, forming a dense plexus of neurites at the inner surface of the retina. These results indicate that massive stimulation of the neuronal growth program can lead to aberrant growth; the absence of local regulatory and guidance factors in the adult, injured optic nerve may therefore represent a major, so far underestimated obstacle to successful axon regeneration.


Assuntos
Fator Neurotrófico Ciliar/genética , Técnicas de Transferência de Genes , Regeneração Nervosa/genética , Traumatismos do Nervo Óptico/genética , Animais , Axônios , Western Blotting , Sobrevivência Celular/genética , Fator Neurotrófico Ciliar/metabolismo , Imunofluorescência , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Artigo em Inglês | MEDLINE | ID: mdl-36376079

RESUMO

Blindness due to rod-cone dystrophies is a significant comorbidity and cause of reduced quality of life worldwide. Optogenetics uses adeno-associated viral (AAV) vectors to bypass lost photoreceptors and transfect remnant cell populations of the degenerated retina aiming to restore vision via the ectopic expression of opsins. The optogenetic targeting of retinal ganglion cells (RGCs) has been remarkably successful and several studies have advanced to clinical trials over the recent years. The inner retina and specifically ON bipolar cells represent even more appealing targets due to their intrinsically coded tasks in parallel processing and fine-tuning of visual signals before reaching the output: RGCs. However, present success with pursuing inner and outer retinal cells for optogenetic vision restoration is limited by multiple factors, including AAV tropism, promoter specificity, and retinal morphofunctional remodeling. Here we provide a review of the evolution of optogenetics, its greatest challenges, and solutions from bench to bedside.


Assuntos
Optogenética , Degeneração Retiniana , Humanos , Invenções , Qualidade de Vida , Células Ganglionares da Retina/metabolismo , Retina/metabolismo , Degeneração Retiniana/metabolismo
20.
Mol Ther ; 19(7): 1212-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21610698

RESUMO

Inherited retinal degeneration results from many different mutations in either photoreceptor-specific or nonphotoreceptor-specific genes. However, nearly all mutations lead to a common blinding phenotype that initiates with rod cell death, followed by loss of cones. In most retinal degenerations, other retinal neuron cell types survive for long periods after blindness from photoreceptor loss. One strategy to restore light responsiveness to a retina rendered blind by photoreceptor degeneration is to express light-regulated ion channels or transporters in surviving retinal neurons. Recent experiments in rodents have restored light-sensitivity by expressing melanopsin or microbial opsins either broadly throughout the retina or selectively in the inner segments of surviving cones or in bipolar cells. Here, we present an approach whereby a genetically and chemically engineered light-gated ionotropic glutamate receptor (LiGluR) is expressed selectively in retinal ganglion cells (RGCs), the longest-surviving cells in retinal blinding diseases. When expressed in the RGCs of a well-established model of retinal degeneration, the rd1 mouse, LiGluR restores light sensitivity to the RGCs, reinstates light responsiveness to the primary visual cortex, and restores both the pupillary reflex and a natural light-avoidance behavior.


Assuntos
Cegueira/terapia , Receptores de Glutamato/metabolismo , Animais , Cegueira/genética , Dependovirus/genética , Eletrorretinografia , Luz , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Glutamato/genética , Células Ganglionares da Retina/metabolismo , Córtex Visual/metabolismo , Córtex Visual/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA