Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Vision Res ; 212: 108311, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37586294

RESUMEN

Usher syndrome type 1B (USH1B) is a deaf-blindness disorder, caused by mutations in the MYO7A gene, which encodes the heavy chain of an unconventional actin-based motor protein. Here, we examined the two retinal isoforms of MYO7A, IF1 and IF2. We compared 3D models of the two isoforms and noted that the 38-amino acid region that is present in IF1 but absent from IF2 affects the C lobe of the FERM1 domain and the opening of a cleft in this potentially important protein binding domain. Expression of each of the two isoforms of human MYO7A and pig and mouse Myo7a was detected in the RPE and neural retina. Quantification by qPCR showed that the expression of IF2 was typically âˆ¼ 7-fold greater than that of IF1. We discuss the implications of these findings for any USH1B gene therapy strategy. Given the current incomplete knowledge of the functions of each isoform, both isoforms should be considered for targeting both the RPE and the neural retina in gene augmentation therapies.


Asunto(s)
Síndromes de Usher , Humanos , Ratones , Animales , Porcinos , Síndromes de Usher/genética , Síndromes de Usher/terapia , Síndromes de Usher/metabolismo , Miosina VIIa/genética , Miosina VIIa/metabolismo , Retina/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Mutación , Terapia Genética
2.
J Clin Invest ; 133(13)2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37227777

RESUMEN

Many patients with diabetic eye disease respond inadequately to anti-VEGF therapies, implicating additional vasoactive mediators in its pathogenesis. We demonstrate that levels of angiogenic proteins regulated by HIF-1 and -2 remain elevated in the eyes of people with diabetes despite treatment with anti-VEGF therapy. Conversely, by inhibiting HIFs, we normalized the expression of multiple vasoactive mediators in mouse models of diabetic eye disease. Accumulation of HIFs and HIF-regulated vasoactive mediators in hyperglycemic animals was observed in the absence of tissue hypoxia, suggesting that targeting HIFs may be an effective early treatment for diabetic retinopathy. However, while the HIF inhibitor acriflavine prevented retinal vascular hyperpermeability in diabetic mice for several months following a single intraocular injection, accumulation of acriflavine in the retina resulted in retinal toxicity over time, raising concerns for its use in patients. Conversely, 32-134D, a recently developed HIF inhibitor structurally unrelated to acriflavine, was not toxic to the retina, yet effectively inhibited HIF accumulation and normalized HIF-regulated gene expression in mice and in human retinal organoids. Intraocular administration of 32-134D prevented retinal neovascularization and vascular hyperpermeability in mice. These results provide the foundation for clinical studies assessing 32-134D for the treatment of patients with diabetic eye disease.


Asunto(s)
Diabetes Mellitus Experimental , Retinopatía Diabética , Neovascularización Retiniana , Humanos , Ratones , Animales , Acriflavina/metabolismo , Acriflavina/farmacología , Acriflavina/uso terapéutico , Diabetes Mellitus Experimental/metabolismo , Retina/metabolismo , Neovascularización Retiniana/metabolismo , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo
3.
Stem Cell Reports ; 18(5): 1138-1154, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37163980

RESUMEN

Human retinal organoid transplantation could potentially be a treatment for degenerative retinal diseases. How the recipient retina regulates the survival, maturation, and proliferation of transplanted organoid cells is unknown. We transplanted human retinal organoid-derived cells into photoreceptor-deficient mice and conducted histology and single-cell RNA sequencing alongside time-matched cultured retinal organoids. Unexpectedly, we observed human cells that migrated into all recipient retinal layers and traveled long distances. Using an unbiased approach, we identified these cells as astrocytes and brain/spinal cord-like neural precursors that were absent or rare in stage-matched cultured organoids. In contrast, retinal progenitor-derived rods and cones remained in the subretinal space, maturing more rapidly than those in the cultured controls. These data suggest that recipient microenvironment promotes the maturation of transplanted photoreceptors while inducing or facilitating the survival of migratory cell populations that are not normally derived from retinal progenitors. These findings have important implications for potential cell-based treatments of retinal diseases.


Asunto(s)
Degeneración Retiniana , Análisis de Expresión Génica de una Sola Célula , Humanos , Ratones , Animales , Diferenciación Celular/fisiología , Retina , Células Fotorreceptoras Retinianas Conos , Degeneración Retiniana/terapia , Organoides/trasplante
5.
Sci Rep ; 12(1): 4009, 2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-35256656

RESUMEN

Inherited retinal disorders and dry age-related macular degeneration are characterized by the degeneration and death of different types of photoreceptors at different rate and locations. Advancement of new therapeutic interventions such as optogenetics gene therapy and cell replacement therapies are dependent on electrophysiological measurements at cellular resolution. Here, we report the development of an optical coherence tomography (OCT) guided micro-focal multi-color laser stimulation and electroretinogram (ERG) platform for highly localized monitoring of retina function. Functional evaluation of wild type and transgenic pigs affected by retinal degeneration was carried out using OCT guided micro-focal ERG (µfERG) with selected stimulation wavelengths for S, M and L cones as well as rod photoreceptors. In wild type pigs, µfERG allowed functional recording from rods and each type of cone photoreceptor cells separately. Furthermore, functional deficits in P23H transgenic pigs consistent with their retinal degeneration phenotype were observed, including decrease in the S and M cone function and lack of rod photoreceptor function. OCT guided µfERG based monitoring of physiological function will enable characterization of animal models of retinal degenerative diseases and evaluation of therapeutic interventions at the cellular level.


Asunto(s)
Degeneración Retiniana , Animales , Animales Modificados Genéticamente , Electrorretinografía , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Degeneración Retiniana/genética , Células Fotorreceptoras Retinianas Bastones/metabolismo , Porcinos , Tomografía de Coherencia Óptica
6.
Sci Adv ; 8(9): eabm1896, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35235351

RESUMEN

For patients with proliferative diabetic retinopathy (PDR) who do not respond adequately to pan-retinal laser photocoagulation (PRP) or anti-vascular endothelial growth factor (VEGF) therapies, we hypothesized that vascular cells within neovascular tissue secrete autocrine/paracrine angiogenic factors that promote disease progression. To identify these factors, we performed multiplex ELISA angiogenesis arrays on aqueous fluid from PDR patients who responded inadequately to anti-VEGF therapy and/or PRP and identified plasminogen activator inhibitor-1 (PAI-1). PAI-1 expression was increased in vitreous biopsies and neovascular tissue from PDR eyes, limited to retinal vascular cells, regulated by the transcription factor hypoxia-inducible factor (HIF)-2α, and necessary and sufficient to stimulate angiogenesis. Using a pharmacologic inhibitor of HIF-2α (PT-2385) or nanoparticle-mediated RNA interference targeting Pai1, we demonstrate that the HIF-2α/PAI-1 axis is necessary for the development of retinal neovascularization in mice. These results suggest that targeting HIF-2α/PAI-1 will be an effective adjunct therapy for the treatment of PDR patients.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Neovascularización Retiniana , Inductores de la Angiogénesis/uso terapéutico , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/etiología , Retinopatía Diabética/metabolismo , Humanos , Ratones , Neovascularización Patológica , Inhibidor 1 de Activador Plasminogénico/genética , Neovascularización Retiniana/etiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Front Cell Dev Biol ; 10: 813538, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35252183

RESUMEN

Transplantation of stem cell-derived retinal pigment epithelium (RPE) cells is a promising potential therapy for currently incurable retinal degenerative diseases like advanced dry age-related macular degeneration. In this study, we designed a set of clinically applicable devices for subretinal implantation of RPE grafts, towards the overarching goal of establishing enabling technologies for cell-based therapeutic approaches to regenerate RPE cells. This RPE transplant kit includes a custom-designed trephine for the production of RPE transplants, a carrier for storage and transportation, and a surgical device for subretinal delivery of RPE transplants. Cell viability assay confirmed biocompatibility of the transplant carrier and high preservation of RPE transplants upon storage and transportation. The transplant surgical device combines foldable technology that minimizes incision size, controlled delivery speed, no fluid reflux, curved translucent tip, usability of loading and in vivo reloading, and ergonomic handle. Furthermore, the complementary design of the transplant carrier and the delivery device resulted in proper grasping, loading, and orientation of the RPE transplants into the delivery device. Proof-of-concept transplantation studies in a porcine model demonstrated no damage or structural change in RPE transplants during surgical manipulation and subretinal deployment. Post-operative assessment confirmed that RPE transplants were delivered precisely, with no damage to the host retina or choroid, and no significant structural change to the RPE transplants. Our novel surgical kit provides a comprehensive set of tools encompassing RPE graft manufacturing to surgical implantation rendering key enabling technologies for pre-clinical and clinical phases of stem cell-derived RPE regenerative therapies.

8.
Mol Neurodegener ; 17(1): 23, 2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35313950

RESUMEN

Across neurodegenerative diseases, common mechanisms may reveal novel therapeutic targets based on neuronal protection, repair, or regeneration, independent of etiology or site of disease pathology. To address these mechanisms and discuss emerging treatments, in April, 2021, Glaucoma Research Foundation, BrightFocus Foundation, and the Melza M. and Frank Theodore Barr Foundation collaborated to bring together key opinion leaders and experts in the field of neurodegenerative disease for a virtual meeting titled "Solving Neurodegeneration". This "think-tank" style meeting focused on uncovering common mechanistic roots of neurodegenerative disease and promising targets for new treatments, catalyzed by the goal of finding new treatments for glaucoma, the world's leading cause of irreversible blindness and the common interest of the three hosting foundations. Glaucoma, which causes vision loss through degeneration of the optic nerve, likely shares early cellular and molecular events with other neurodegenerative diseases of the central nervous system. Here we discuss major areas of mechanistic overlap between neurodegenerative diseases of the central nervous system: neuroinflammation, bioenergetics and metabolism, genetic contributions, and neurovascular interactions. We summarize important discussion points with emphasis on the research areas that are most innovative and promising in the treatment of neurodegeneration yet require further development. The research that is highlighted provides unique opportunities for collaboration that will lead to efforts in preventing neurodegeneration and ultimately vision loss.


Asunto(s)
Glaucoma , Enfermedades Neurodegenerativas , Glaucoma/patología , Humanos , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia , Neuroprotección , Nervio Óptico/patología
9.
Int J Ophthalmol ; 15(1): 157-166, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35047371

RESUMEN

Dry age-related macular degeneration (AMD) is a progressive blinding disease that currently affects millions of people worldwide with no successful treatment available. Significant research efforts are currently underway to develop therapies aimed at slowing the progression of this disease or, more notably, reversing it. Here the therapies which have reached clinical trial for treatment of dry AMD were reviewed. A thorough search of PubMed, Embase, and Clinicaltrials.gov has led to a comprehensive collection of the most recent strategies being evaluated. This review also endeavors to assess the status and future directions of therapeutics for this debilitating condition.

10.
J Extracell Vesicles ; 10(13): e12165, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34750957

RESUMEN

Age-related macular degeneration (AMD) is a leading cause of blindness worldwide. Drusen are key contributors to the etiology of AMD and the ability to modulate drusen biogenesis could lead to therapeutic strategies to slow or halt AMD progression. The mechanisms underlying drusen biogenesis, however, remain mostly unknown. Here we demonstrate that under homeostatic conditions extracellular vesicles (EVs) secreted by retinal pigment epithelium (RPE) cells are enriched in proteins associated with mechanisms involved in AMD pathophysiology, including oxidative stress, immune response, inflammation, complement system and drusen composition. Furthermore, we provide first evidence that drusen-associated proteins are released as cargo of extracellular vesicles secreted by RPE cells in a polarised apical:basal mode. Notably, drusen-associated proteins exhibited distinctive directional secretion modes in homeostatic conditions and, differential modulation of this directional secretion in response to AMD stressors. These observations underpin the existence of a finely-tuned mechanism regulating directional apical:basal sorting and secretion of drusen-associated proteins via EVs, and its modulation in response to mechanisms involved in AMD pathophysiology. Collectively, our results strongly support an active role of RPE-derived EVs as a key source of drusen proteins and important contributors to drusen development and growth.


Asunto(s)
Polaridad Celular/efectos de los fármacos , Vesículas Extracelulares/metabolismo , Degeneración Macular/complicaciones , Degeneración Macular/metabolismo , Proteínas/metabolismo , Drusas Retinianas/complicaciones , Drusas Retinianas/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal/efectos de los fármacos , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Nicotina/farmacología , Organoides/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fagocitosis , Especies Reactivas de Oxígeno/metabolismo , Secretoma/metabolismo
11.
Transl Vis Sci Technol ; 10(10): 9, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34383873

RESUMEN

Translational Relevance: Designing a GMP-compliant protocol for three-dimensional retinal organoid production is of urgent need in order to bring transplantation of hiPSC-derived retinal tissue and derived cells to clinical trials - and ultimately patient treatment - for retinal degenerative diseases.


Asunto(s)
Células Madre Pluripotentes Inducidas , Degeneración Retiniana , Humanos , Organoides , Retina/cirugía
12.
J Clin Invest ; 131(12)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34128478

RESUMEN

Therapies targeting VEGF have proven only modestly effective for the treatment of proliferative sickle cell retinopathy (PSR), the leading cause of blindness in patients with sickle cell disease. Here, we shift our attention upstream from the genes that promote retinal neovascularization (NV) to the transcription factors that regulate their expression. We demonstrated increased expression of HIF-1α and HIF-2α in the ischemic inner retina of PSR eyes. Although both HIFs participated in promoting VEGF expression by hypoxic retinal Müller cells, HIF-1 alone was sufficient to promote retinal NV in mice, suggesting that therapies targeting only HIF-2 would not be adequate to prevent PSR. Nonetheless, administration of a HIF-2-specific inhibitor currently in clinical trials (PT2385) inhibited NV in the oxygen-induced retinopathy (OIR) mouse model. To unravel these discordant observations, we examined the expression of HIFs in OIR mice and demonstrated rapid but transient accumulation of HIF-1α but delayed and sustained accumulation of HIF-2α; simultaneous expression of HIF-1α and HIF-2α was not observed. Staggered HIF expression was corroborated in hypoxic adult mouse retinal explants but not in human retinal organoids, suggesting that this phenomenon may be unique to mice. Using pharmacological inhibition or an in vivo nanoparticle-mediated RNAi approach, we demonstrated that inhibiting either HIF was effective for preventing NV in OIR mice. Collectively, these results explain why inhibition of either HIF-1α or HIF-2α is equally effective for preventing retinal NV in mice but suggest that therapies targeting both HIFs will be necessary to prevent NV in patients with PSR.


Asunto(s)
Anemia de Células Falciformes/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Neovascularización Retiniana/metabolismo , Vasos Retinianos/metabolismo , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Modelos Animales de Enfermedad , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Indanos/farmacología , Ratones , Neovascularización Retiniana/etiología , Neovascularización Retiniana/genética , Sulfonas/farmacología
13.
Front Immunol ; 12: 621007, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054796

RESUMEN

Replacement of dysfunctional retinal pigmented epithelium (RPE) with grafts derived from stem cells has the potential to improve vision for patients with retinal disorders. In fact, the potential is such that a great number of groups are attempting to realize this therapy through individual strategies with a variety of stem cell products, hosts, immunomodulatory regimen, and techniques to assess the success of their design. Comparing the findings of different investigators is complicated by a number of factors. The immune response varies greatly between xenogeneic and allogeneic transplantation. A unique immunologic environment is created in the subretinal space, the target of RPE grafts. Both functional assessment and imaging techniques used to evaluate transplants are susceptible to erroneous conclusions. Lastly, the pharmacologic regimens used in RPE transplant trials are as numerous and variable as the trials themselves, making it difficult to determine useful results. This review will discuss the causes of these complicating factors, digest the strategies and results from clinical and preclinical studies, and suggest places for improvement in the design of future transplants and investigations.


Asunto(s)
Rechazo de Injerto/inmunología , Células Madre Pluripotentes Inducidas/fisiología , Degeneración Macular/terapia , Trasplante de Órganos , Epitelio Pigmentado de la Retina/fisiología , Animales , Humanos , Epitelio Pigmentado de la Retina/trasplante , Tolerancia al Trasplante
14.
Prog Retin Eye Res ; 75: 100779, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31494256

RESUMEN

Stem cell transplantation holds great promise as a potential treatment for currently incurable retinal degenerative diseases that cause poor vision and blindness. Recently, safety data have emerged from several Phase I/II clinical trials of retinal stem cell transplantation. These clinical trials, usually run in partnership with academic institutions, are based on sound preclinical studies and are focused on patient safety. However, reports of serious adverse events arising from cell therapy in other poorly regulated centers have now emerged in the lay and scientific press. While progress in stem cell research for blindness has been greeted with great enthusiasm by patients, scientists, doctors and industry alike, these adverse events have raised concerns about the safety of retinal stem cell transplantation and whether patients are truly protected from undue harm. The aim of this review is to summarize and appraise the safety of human retinal stem cell transplantation in the context of its potential to be developed into an effective treatment for retinal degenerative diseases.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Pluripotentes Inducidas/trasplante , Retina/citología , Degeneración Retiniana/terapia , Epitelio Pigmentado de la Retina/trasplante , Trasplante de Células Madre/métodos , Humanos
15.
Brain Pathol ; 29(1): 45-52, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29668072

RESUMEN

We have identified a discrete, focal telomere DNA expansion phenotype in the photoreceptor cell layer of normal, non-neoplastic human retinas. This phenotype is similar to that observed in a subset of human cancers, including a large fraction of tumors of the central nervous system, which maintain their telomeres via the non-telomerase-mediated alternative lengthening of telomeres (ALT) mechanism. We observed that these large, ultra-bright telomere DNA foci are restricted to the rod photoreceptors and are not observed in other cell types. Additionally, focus-positive rod cells are dispersed homogeneously throughout the posterior retinal photoreceptor cell layer and appear to be human-specific. We examined 108 normal human retinas obtained at autopsy from a wide range of ages. These large, ultra-bright telomere DNA foci were not observed in infants before 6 months of age; however, the prevalence of focus-positive rod cells dramatically increased throughout life. To investigate associations between this phenotype and retinal pathology, we assessed adult glaucoma (N = 29) and diabetic retinopathy (N = 38) cases. Focus-positive rod cells were prominent in these diseases. When compared to the normal group, after adjusting for age, logistic regression modeling revealed significantly increased odds of falling in the high category of focus-positive rod cells for glaucoma and diabetic retinopathy. In summary, we have identified a dramatic telomere alteration associated with aging and diseases affecting the retina.


Asunto(s)
Células Fotorreceptoras Retinianas Bastones/fisiología , Homeostasis del Telómero/genética , Telómero/genética , Factores de Edad , Envejecimiento/genética , Animales , ADN , Retinopatía Diabética/genética , Retinopatía Diabética/fisiopatología , Femenino , Glaucoma/genética , Glaucoma/fisiopatología , Humanos , Masculino , Fenotipo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/fisiología , Retina/fisiología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Telómero/fisiología
17.
Sci Rep ; 8(1): 2823, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29434302

RESUMEN

A range of cell types, including embryonic stem cells, neurons and astrocytes have been shown to release extracellular vesicles (EVs) containing molecular cargo. Across cell types, EVs facilitate transfer of mRNA, microRNA and proteins between cells. Here we describe the release kinetics and content of EVs from mouse retinal progenitor cells (mRPCs). Interestingly, mRPC derived EVs contain mRNA, miRNA and proteins associated with multipotency and retinal development. Transcripts enclosed in mRPC EVs, include the transcription factors Pax6, Hes1, and Sox2, a mitotic chromosome stabilizer Ki67, and the neural intermediate filaments Nestin and GFAP. Proteomic analysis of EV content revealed retinogenic growth factors and morphogen proteins. mRPC EVs were shown to transfer GFP mRNA between cell populations. Finally, analysis of EV mediated functional cargo delivery, using the Cre-loxP recombination system, revealed transfer and uptake of Cre+ EVs, which were then internalized by target mRPCs activating responder loxP GFP expression. In summary, the data supports a paradigm of EV genetic material encapsulation and transfer within RPC populations. RPC EV transfer may influence recipient RPC transcriptional and post-transcriptional regulation, representing a novel mechanism of differentiation and fate determination during retinal development.


Asunto(s)
Vesículas Extracelulares/metabolismo , Retina/metabolismo , Células Madre/metabolismo , Animales , Astrocitos/metabolismo , Diferenciación Celular , Células Cultivadas , Vesículas Extracelulares/fisiología , Regulación de la Expresión Génica/genética , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , MicroARNs/genética , Neuronas/metabolismo , Proteómica , ARN Mensajero/metabolismo , Retina/fisiología , Factores de Transcripción/metabolismo
18.
Development ; 144(20): 3698-3705, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28870990

RESUMEN

The advent of stem cell-derived retinal organoids has brought forth unprecedented opportunities for developmental and physiological studies, while presenting new therapeutic promise for retinal degenerative diseases. From a translational perspective, organoid systems provide exciting new prospects for drug discovery, offering the possibility to perform compound screening in a three-dimensional (3D) human tissue context that resembles the native histoarchitecture and to some extent recapitulates cellular interactions. However, inherent variability issues and a general lack of robust quantitative technologies for analyzing organoids on a large scale pose severe limitations for their use in translational applications. To address this need, we have developed a screening platform that enables accurate quantification of fluorescent reporters in complex human iPSC-derived retinal organoids. This platform incorporates a fluorescence microplate reader that allows xyz-dimensional detection and fine-tuned wavelength selection. We have established optimal parameters for fluorescent reporter signal detection, devised methods to compensate for organoid size variability, evaluated performance and sensitivity parameters, and validated this technology for functional applications.


Asunto(s)
Técnicas Genéticas , Células Madre Pluripotentes Inducidas/citología , Organoides/fisiología , Retina/fisiología , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular , Colorantes Fluorescentes , Genes Reporteros , Humanos , Microscopía Fluorescente , Estrés Oxidativo , Trasplante de Células Madre , Transgenes , Investigación Biomédica Traslacional
19.
Development ; 143(23): 4368-4380, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27660325

RESUMEN

The derivation and maintenance of human pluripotent stem cells (hPSCs) in stable naïve pluripotent states has a wide impact in human developmental biology. However, hPSCs are unstable in classical naïve mouse embryonic stem cell (ESC) WNT and MEK/ERK signal inhibition (2i) culture. We show that a broad repertoire of conventional hESC and transgene-independent human induced pluripotent stem cell (hiPSC) lines could be reverted to stable human preimplantation inner cell mass (ICM)-like naïve states with only WNT, MEK/ERK, and tankyrase inhibition (LIF-3i). LIF-3i-reverted hPSCs retained normal karyotypes and genomic imprints, and attained defining mouse ESC-like functional features, including high clonal self-renewal, independence from MEK/ERK signaling, dependence on JAK/STAT3 and BMP4 signaling, and naïve-specific transcriptional and epigenetic configurations. Tankyrase inhibition promoted a stable acquisition of a human preimplantation ICM-like ground state via modulation of WNT signaling, and was most efficacious in efficiently reprogrammed conventional hiPSCs. Importantly, naïve reversion of a broad repertoire of conventional hiPSCs reduced lineage-primed gene expression and significantly improved their multilineage differentiation capacities. Stable naïve hPSCs with reduced genetic variability and improved functional pluripotency will have great utility in regenerative medicine and human disease modeling.


Asunto(s)
Diferenciación Celular/fisiología , Autorrenovación de las Células/fisiología , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Tanquirasas/antagonistas & inhibidores , Vía de Señalización Wnt/fisiología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Células Cultivadas , Reprogramación Celular/fisiología , Estratos Germinativos/embriología , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Humanos , Quinasas Janus/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Ratones , Factor de Transcripción STAT3/metabolismo
20.
J Vis Exp ; (105): e52002, 2015 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-26556302

RESUMEN

The cone photoreceptor-enriched cultures derived from embryonic chick retinas have become an indispensable tool for researchers around the world studying the biology of retinal neurons, particularly photoreceptors. The applications of this system go beyond basic research, as they can easily be adapted to high throughput technologies for drug development. However, genetic manipulation of retinal photoreceptors in these cultures has proven to be very challenging, posing an important limitation to the usefulness of the system. We have recently developed and validated an ex ovo plasmid electroporation technique that increases the rate of transfection of retinal cells in these cultures by five-fold compared to other currently available protocols(1). In this method embryonic chick eyes are enucleated at stage 27, the RPE is removed, and the retinal cup is placed in a plasmid-containing solution and electroporated using easily constructed custom-made electrodes. The retinas are then dissociated and cultured using standard procedures. This technique can be applied to overexpression studies as well as to the downregulation of gene expression, for example via the use of plasmid-driven RNAi technology, commonly achieving transgene expression in 25% of the photoreceptor population. The video format of the present publication will make this technology easily accessible to researchers in the field, enabling the study of gene function in primary retinal cultures. We have also included detailed explanations of the critical steps of this procedure for a successful outcome and reproducibility.


Asunto(s)
Electroporación/métodos , Retina/fisiología , Transfección/métodos , Animales , Embrión de Pollo , Células Fotorreceptoras de Vertebrados/citología , Células Fotorreceptoras de Vertebrados/fisiología , Plásmidos/administración & dosificación , Plásmidos/genética , Cultivo Primario de Células , Reproducibilidad de los Resultados , Retina/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA