Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Med ; 27(7): 1223-1229, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34031601

RESUMO

Optogenetics may enable mutation-independent, circuit-specific restoration of neuronal function in neurological diseases. Retinitis pigmentosa is a neurodegenerative eye disease where loss of photoreceptors can lead to complete blindness. In a blind patient, we combined intraocular injection of an adeno-associated viral vector encoding ChrimsonR with light stimulation via engineered goggles. The goggles detect local changes in light intensity and project corresponding light pulses onto the retina in real time to activate optogenetically transduced retinal ganglion cells. The patient perceived, located, counted and touched different objects using the vector-treated eye alone while wearing the goggles. During visual perception, multichannel electroencephalographic recordings revealed object-related activity above the visual cortex. The patient could not visually detect any objects before injection with or without the goggles or after injection without the goggles. This is the first reported case of partial functional recovery in a neurodegenerative disease after optogenetic therapy.


Assuntos
Cegueira/fisiopatologia , Cegueira/terapia , Terapia Genética/métodos , Optogenética/métodos , Retinose Pigmentar/patologia , Ondas Encefálicas/fisiologia , Dependovirus/genética , Dispositivos de Proteção dos Olhos , Vetores Genéticos/genética , Humanos , Masculino , Pessoa de Meia-Idade , Células Fotorreceptoras/fisiologia , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/fisiologia , Visão Ocular/fisiologia , Córtex Visual/fisiologia , Percepção Visual/fisiologia
2.
Front Cell Neurosci ; 15: 648210, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33815066

RESUMO

Human-induced pluripotent stem cell (hiPSC) derived organoids have become increasingly used systems allowing 3D-modeling of human organ development, and disease. They are also a reliable source of cells for transplantation in cell therapy and an excellent model to validate gene therapies. To make full use of these systems, a toolkit of genetic modification techniques is necessary to control their activity in line with the downstream application. We have previously described adeno-associated viruse (AAV) vectors for efficient targeting of cells within human retinal organoids. Here, we describe biological restriction and enhanced gene expression in cone cells of such organoids thanks to the use of a 1.7-kb L-opsin promoter. We illustrate the usefulness of implementing such a promoter to enhance the expression of the red-shifted opsin Jaws in fusion with a fluorescent reporter gene, enabling cell sorting to enrich the desired cell population. Increased Jaws expression after transplantation improved light responses promising better therapeutic outcomes in a cell therapy setting. Our results point to the importance of promoter activity in restricting, improving, and controlling the kinetics of transgene expression during the maturation of hiPSC retinal derivatives. Differentiation requires mechanisms to initiate specific transcriptional changes and to reinforce those changes when mature cell states are reached. By employing a cell-type-specific promoter we put transgene expression under the new transcriptional program of mature cells.

3.
Front Cell Dev Biol ; 8: 585675, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33195235

RESUMO

Optic neuropathies are a major cause of visual impairment due to retinal ganglion cell (RGC) degeneration. Human induced-pluripotent stem cells (iPSCs) represent a powerful tool for studying both human RGC development and RGC-related pathological mechanisms. Because RGC loss can be massive before the diagnosis of visual impairment, cell replacement is one of the most encouraging strategies. The present work describes the generation of functional RGCs from iPSCs based on innovative 3D/2D stepwise differentiation protocol. We demonstrate that targeting the cell surface marker THY1 is an effective strategy to select transplantable RGCs. By generating a fluorescent GFP reporter iPSC line to follow transplanted cells, we provide evidence that THY1-positive RGCs injected into the vitreous of mice with optic neuropathy can survive up to 1 month, intermingled with the host RGC layer. These data support the usefulness of iPSC-derived RGC exploration as a potential future therapeutic strategy for optic nerve regeneration.

4.
Cell Rep ; 33(1): 108220, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33027659

RESUMO

Axonal arbors in many neuronal networks are exuberant early during development and become refined by activity-dependent competitive mechanisms. Theoretical work proposed non-competitive interactions between co-active axons to co-stabilize their connections, but the demonstration of such interactions is lacking. Here, we provide experimental evidence that reducing cyclic AMP (cAMP) signaling in a subset of retinal ganglion cells favors the elimination of thalamic projections from neighboring neurons, pointing to a cAMP-dependent interaction that promotes axon stabilization.


Assuntos
Axônios/metabolismo , AMP Cíclico/metabolismo , Neurônios/metabolismo , Humanos , Transdução de Sinais
5.
Int J Mol Sci ; 21(3)2020 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-32028585

RESUMO

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.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Organoides/metabolismo , Retina/metabolismo , Animais , Terapia Genética , Variação Genética , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Organoides/citologia , Receptores de Superfície Celular/metabolismo , Retina/citologia , Transdução Genética , Transgenes
6.
Nat Commun ; 10(1): 4524, 2019 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-31586094

RESUMO

A major challenge in the treatment of retinal degenerative diseases, with the transplantation of replacement photoreceptors, is the difficulty in inducing the grafted cells to grow and maintain light sensitive outer segments in the host retina, which depends on proper interaction with the underlying retinal pigment epithelium (RPE). Here, for an RPE-independent treatment approach, we introduce a hyperpolarizing microbial opsin into photoreceptor precursors from newborn mice, and transplant them into blind mice lacking the photoreceptor layer. These optogenetically-transformed photoreceptors are light responsive and their transplantation leads to the recovery of visual function, as shown by ganglion cell recordings and behavioral tests. Subsequently, we generate cone photoreceptors from human induced pluripotent stem cells, expressing the chloride pump Jaws. After transplantation into blind mice, we observe light-driven responses at the photoreceptor and ganglion cell levels. These results demonstrate that structural and functional retinal repair is possible by combining stem cell therapy and optogenetics.


Assuntos
Engenharia Celular/métodos , Optogenética/métodos , Células Fotorreceptoras de Vertebrados/transplante , Degeneração Retiniana/terapia , Animais , Animais Recém-Nascidos , Técnicas de Cultura de Células/métodos , Dependovirus/genética , Modelos Animais de Doenças , Feminino , Vetores Genéticos/genética , Células HEK293 , Halorrodopsinas/genética , Humanos , Células-Tronco Pluripotentes Induzidas , Masculino , Camundongos , Camundongos Knockout , Degeneração Retiniana/genética , Rodopsina/genética , Transfecção , Resultado do Tratamento
7.
Stem Cell Reports ; 11(3): 665-680, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30100409

RESUMO

Photoreceptor degenerative diseases are a major cause of blindness for which cell replacement is one of the most encouraging strategies. For stem cell-based therapy using human induced pluripotent stem cells (hiPSCs), it is crucial to obtain a homogenous photoreceptor cell population. We confirmed that the cell surface antigen CD73 is exclusively expressed in hiPSC-derived photoreceptors by generating a fluorescent cone rod homeobox (Crx) reporter hiPSC line using CRISPR/Cas9 genome editing. We demonstrated that CD73 targeting by magnetic-activated cell sorting (MACS) is an effective strategy to separate a safe population of transplantable photoreceptors. CD73+ photoreceptor precursors can be isolated in large numbers and transplanted into rat eyes, showing capacity to survive and mature in close proximity to host inner retina of a model of photoreceptor degeneration. These data demonstrate that CD73+ photoreceptor precursors hold great promise for a future safe clinical translation.


Assuntos
5'-Nucleotidase/análise , Células-Tronco Pluripotentes Induzidas/citologia , Organoides/citologia , Retina/citologia , Células Fotorreceptoras Retinianas Bastonetes/citologia , Animais , Linhagem Celular , Proteínas Ligadas por GPI/análise , Humanos , Organoides/transplante , Ratos Nus , Células Fotorreceptoras Retinianas Bastonetes/transplante
8.
Adv Exp Med Biol ; 1074: 69-73, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29721929

RESUMO

The expression of light-sensitive microbial opsins is a promising mutation-independent approach to restore vision in retinal degenerative diseases. Using viral vectors, optogenetic tools can be genetically expressed in various subpopulations of retinal neurons. The choice of cell type depends on the availability of surviving retinal cells. If cones are still alive but they lack outer segments, they can be targeted with optogenetic inhibitors, such as halorhodopsin. Alternatively, it is possible to bypass the photoreceptors and to target bipolar cells. In late-stage degeneration, when bipolar cells degenerate, "artificial photoreceptors" can be made from retinal ganglion cells, but with this approach, upstream retinal processing cannot be utilized. However, when ganglion cells are stimulated directly, higher brain regions might be able to compensate for some loss of retinal processing, which is indicated by clinical studies with epiretinal implants, where patients can perform simple visual tasks. Finally, optogenetics in combination with neuroprotective approaches could serve as a valuable strategy to restore the function of remaining cells, as well as to rescue retinal neurons from progressive degeneration.


Assuntos
Vetores Genéticos/uso terapêutico , Optogenética/métodos , Degeneração Retiniana/terapia , Rodopsinas Microbianas/uso terapêutico , Células Amácrinas/fisiologia , Dependovirus/genética , Humanos , Fármacos Neuroprotetores/uso terapêutico , Especificidade de Órgãos , Células Bipolares da Retina/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Células Ganglionares da Retina/fisiologia , Células Ganglionares da Retina/efeitos da radiação , Rodopsinas Microbianas/genética , Próteses Visuais
9.
JCI Insight ; 3(2)2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29367457

RESUMO

Intraocular injection of adeno-associated viral (AAV) vectors has been an evident route for delivering gene drugs into the retina. However, gaps in our understanding of AAV transduction patterns within the anatomically unique environments of the subretinal and intravitreal space of the primate eye impeded the establishment of noninvasive and efficient gene delivery to foveal cones in the clinic. Here, we establish new vector-promoter combinations to overcome the limitations associated with AAV-mediated cone transduction in the fovea with supporting studies in mouse models, human induced pluripotent stem cell-derived organoids, postmortem human retinal explants, and living macaques. We show that an AAV9 variant provides efficient foveal cone transduction when injected into the subretinal space several millimeters away from the fovea, without detaching this delicate region. An engineered AAV2 variant provides gene delivery to foveal cones with a well-tolerated dose administered intravitreally. Both delivery modalities rely on a cone-specific promoter and result in high-level transgene expression compatible with optogenetic vision restoration. The model systems described here provide insight into the behavior of AAV vectors across species to obtain safety and efficacy needed for gene therapy in neurodegenerative disorders.


Assuntos
Fóvea Central/patologia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Transdução Genética/métodos , Transtornos da Visão/terapia , Animais , Linhagem Celular , Dependovirus/genética , Feminino , Fóvea Central/diagnóstico por imagem , Vetores Genéticos/genética , Humanos , Células-Tronco Pluripotentes Induzidas , Injeções Intraoculares , Microscopia Intravital , Macaca fascicularis , Masculino , Camundongos , Modelos Animais , Optogenética/métodos , Técnicas de Patch-Clamp , Regiões Promotoras Genéticas/genética , Transgenes/genética , Transtornos da Visão/genética , Transtornos da Visão/patologia
10.
Mol Ther ; 25(11): 2546-2560, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28807567

RESUMO

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.


Assuntos
Channelrhodopsins/genética , Vetores Genéticos/administração & dosagem , Regiões Promotoras Genéticas , Recuperação de Função Fisiológica , Degeneração Retiniana/terapia , Animais , Channelrhodopsins/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Injeções Intravítreas , Luz , Macaca fascicularis , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Optogenética , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Transdução Genética , Transgenes , Visão Ocular/fisiologia
11.
Stem Cells ; 35(5): 1176-1188, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28220575

RESUMO

Human induced pluripotent stem cells (hiPSCs) are potentially useful in regenerative therapies for retinal disease. For medical applications, therapeutic retinal cells, such as retinal pigmented epithelial (RPE) cells or photoreceptor precursors, must be generated under completely defined conditions. To this purpose, we have developed a two-step xeno-free/feeder-free (XF/FF) culture system to efficiently differentiate hiPSCs into retinal cells. This simple method, relies only on adherent hiPSCs cultured in chemically defined media, bypassing embryoid body formation. In less than 1 month, adherent hiPSCs are able to generate self-forming neuroretinal-like structures containing retinal progenitor cells (RPCs). Floating cultures of isolated structures enabled the differentiation of RPCs into all types of retinal cells in a sequential overlapping order, with the generation of transplantation-compatible CD73+ photoreceptor precursors in less than 100 days. Our XF/FF culture conditions allow the maintenance of both mature cones and rods in retinal organoids until 280 days with specific photoreceptor ultrastructures. Moreover, both hiPSC-derived retinal organoids and dissociated retinal cells can be easily cryopreserved while retaining their phenotypic characteristics and the preservation of CD73+ photoreceptor precursors. Concomitantly to neural retina, this process allows the generation of RPE cells that can be effortlessly amplified, passaged, and frozen while retaining a proper RPE phenotype. These results demonstrate that simple and efficient retinal differentiation of adherent hiPSCs can be accomplished in XF/FF conditions. This new method is amenable to the development of an in vitro GMP-compliant retinal cell manufacturing protocol allowing large-scale production and banking of hiPSC-derived retinal cells and tissues. Stem Cells 2017;35:1176-1188.


Assuntos
Células Alimentadoras/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Organoides/citologia , Preservação Biológica , Epitélio Pigmentado da Retina/citologia , Adesão Celular , Diferenciação Celular , Linhagem Celular , Criopreservação , Humanos , Organoides/ultraestrutura , Células Fotorreceptoras/citologia
12.
EMBO Mol Med ; 8(11): 1248-1264, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27679671

RESUMO

Targeting the photosensitive ion channel channelrhodopsin-2 (ChR2) to the retinal circuitry downstream of photoreceptors holds promise in treating vision loss caused by retinal degeneration. However, the high intensity of blue light necessary to activate channelrhodopsin-2 exceeds the safety threshold of retinal illumination because of its strong potential to induce photochemical damage. In contrast, the damage potential of red-shifted light is vastly lower than that of blue light. Here, we show that a red-shifted channelrhodopsin (ReaChR), delivered by AAV injections in blind rd1 mice, enables restoration of light responses at the retinal, cortical, and behavioral levels, using orange light at intensities below the safety threshold for the human retina. We further show that postmortem macaque retinae infected with AAV-ReaChR can respond with spike trains to orange light at safe intensities. Finally, to directly address the question of translatability to human subjects, we demonstrate for the first time, AAV- and lentivirus-mediated optogenetic spike responses in ganglion cells of the postmortem human retina.


Assuntos
Terapia Genética/métodos , Fototerapia/métodos , Retina/fisiologia , Degeneração Retiniana/terapia , Rodopsina/genética , Animais , Dependovirus/genética , Vetores Genéticos , Humanos , Lentivirus/genética , Luz , Macaca , Camundongos , Rodopsina/metabolismo , Transdução Genética , Resultado do Tratamento
13.
Mol Ther ; 23(1): 7-16, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25095892

RESUMO

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.


Assuntos
Cegueira/terapia , Dependovirus/genética , Terapia Genética/métodos , Células Bipolares da Retina/metabolismo , Degeneração Retiniana/terapia , Animais , Comportamento Animal , Cegueira/genética , Cegueira/patologia , Channelrhodopsins , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Engenharia Genética , Vetores Genéticos , Injeções Intravítreas , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas , Células Bipolares da Retina/patologia , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Percepção Visual/genética , Corpo Vítreo
14.
Curr Opin Neurol ; 28(1): 51-60, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25545056

RESUMO

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.


Assuntos
Proteínas do Olho/genética , Terapia Genética/métodos , Mutação , Doenças Retinianas/terapia , Dependovirus , Vetores Genéticos , Humanos , Amaurose Congênita de Leber/genética , Amaurose Congênita de Leber/terapia , Degeneração Retiniana/genética , Degeneração Retiniana/terapia , Doenças Retinianas/genética
15.
Science ; 329(5990): 413-7, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20576849

RESUMO

Retinitis pigmentosa refers to a diverse group of hereditary diseases that lead to incurable blindness, affecting two million people worldwide. As a common pathology, rod photoreceptors die early, whereas light-insensitive, morphologically altered cone photoreceptors persist longer. It is unknown if these cones are accessible for therapeutic intervention. Here, we show that expression of archaebacterial halorhodopsin in light-insensitive cones can substitute for the native phototransduction cascade and restore light sensitivity in mouse models of retinitis pigmentosa. Resensitized photoreceptors activate all retinal cone pathways, drive sophisticated retinal circuit functions (including directional selectivity), activate cortical circuits, and mediate visually guided behaviors. Using human ex vivo retinas, we show that halorhodopsin can reactivate light-insensitive human photoreceptors. Finally, we identified blind patients with persisting, light-insensitive cones for potential halorhodopsin-based therapy.


Assuntos
Terapia Genética , Halorrodopsinas/genética , Halorrodopsinas/metabolismo , Células Fotorreceptoras Retinianas Cones/fisiologia , Retinose Pigmentar/terapia , Animais , Dependovirus/genética , Modelos Animais de Doenças , Potenciais Evocados Visuais , Vetores Genéticos , Halobacteriaceae/genética , Humanos , Luz , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas , Retina/fisiologia , Células Ganglionares da Retina/fisiologia , Retinose Pigmentar/fisiopatologia , Técnicas de Cultura de Tecidos , Transfecção , Visão Ocular , Vias Visuais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA