Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomed Opt Express ; 13(6): 3476-3492, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35781966

RESUMEN

Photoreceptors are the key functional cell types responsible for the initiation of vision in the retina. Phototransduction involves isomerization and conversion of vitamin A compounds, known as retinoids, and their recycling through the visual cycle. We demonstrate a functional readout of the visual cycle in photoreceptors within stem cell-derived retinal organoids and mouse retinal explants based on spectral and lifetime changes in autofluorescence of the visual cycle retinoids after exposure to light or chemical stimuli. We also apply a simultaneous two- and three-photon excitation method that provides specific signals and increases contrast between these retinoids, allowing for reliable detection of their presence and conversion within photoreceptors. This multiphoton imaging technique resolves the slow dynamics of visual cycle reactions and can enable high-throughput functional screening of retinal tissues and organoid cultures with single-cell resolution.

2.
Commun Biol ; 3(1): 82, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-32081919

RESUMEN

Photoreceptor loss is a leading cause of blindness, but mechanisms underlying photoreceptor degeneration are not well understood. Treatment strategies would benefit from improved understanding of gene-expression patterns directing photoreceptor development, as many genes are implicated in both development and degeneration. Neural retina leucine zipper (NRL) is critical for rod photoreceptor genesis and degeneration, with NRL mutations known to cause enhanced S-cone syndrome and retinitis pigmentosa. While murine Nrl loss has been characterized, studies of human NRL can identify important insights for human retinal development and disease. We utilized iPSC organoid models of retinal development to molecularly define developmental alterations in a human model of NRL loss. Consistent with the function of NRL in rod fate specification, human retinal organoids lacking NRL develop S-opsin dominant photoreceptor populations. We report generation of two distinct S-opsin expressing populations in NRL null retinal organoids and identify MEF2C as a candidate regulator of cone development.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Proteínas del Ojo/genética , Células Madre Pluripotentes Inducidas/fisiología , Organoides/patología , Retina/patología , Células Fotorreceptoras Retinianas Conos/fisiología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Estudios de Casos y Controles , Diferenciación Celular/genética , Células Cultivadas , Reprogramación Celular/fisiología , Enfermedades Hereditarias del Ojo/genética , Enfermedades Hereditarias del Ojo/patología , Feto/patología , Perfilación de la Expresión Génica , Humanos , Regeneración Nerviosa/genética , Neurogénesis/genética , Organoides/fisiología , Cultivo Primario de Células/métodos , Retina/fisiología , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Transcriptoma , Trastornos de la Visión/genética , Trastornos de la Visión/patología
3.
Hum Mol Genet ; 29(6): 967-979, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32011687

RESUMEN

Inherited retinal degenerations (IRDs) are at the focus of current genetic therapeutic advancements. For a genetic treatment such as gene therapy to be successful, an accurate genetic diagnostic is required. Genetic diagnostics relies on the assessment of the probability that a given DNA variant is pathogenic. Non-coding variants present a unique challenge for such assessments as compared to coding variants. For one, non-coding variants are present at much higher number in the genome than coding variants. In addition, our understanding of the rules that govern the non-coding regions of the genome is less complete than our understanding of the coding regions. Methods that allow for both the identification of candidate non-coding pathogenic variants and their functional validation may help overcome these caveats allowing for a greater number of patients to benefit from advancements in genetic therapeutics. We present here an unbiased approach combining whole genome sequencing (WGS) with patient-induced pluripotent stem cell (iPSC)-derived retinal organoids (ROs) transcriptome analysis. With this approach, we identified and functionally validated a novel pathogenic non-coding variant in a small family with a previously unresolved genetic diagnosis.


Asunto(s)
Marcadores Genéticos , Variación Genética , Genoma Humano , RNA-Seq/métodos , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Secuenciación Completa del Genoma/métodos , Niño , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Linaje , Secuenciación del Exoma
4.
Am J Ophthalmol ; 206: 113-131, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31078532

RESUMEN

PURPOSE: To investigate the role of fibroblast growth factors (FGFs) in the production of neural retina (NR) and retinal pigmented epithelium (RPE) in a human pluripotent stem cell model of early retinal development. METHODS: Human induced pluripotent stem cell (hiPSC) lines from an individual with microphthalmia caused by a functional null mutation (R200Q) in visual system homeobox 2 (VSX2), a transcription factor involved in early NR progenitor cell (NRPC) production, and a normal sibling were differentiated along the retinal and forebrain lineages using an established protocol. Quantitative and global gene expression analyses (microarray and RNAseq) were used to investigate endogenous FGF expression profiles in these cultures over time. Based on these results, mutant and control hiPSC cultures were treated exogenously with selected FGFs and subjected to gene and protein expression analyses to determine their effects on RPE and NR production. RESULTS: We found that FGF9 and FGF19 were selectively increased in early hiPSC-derived optic vesicles (OVs) when compared to isogenic cultures of hiPSC-derived forebrain neurospheres. Furthermore, these same FGFs were downregulated over time in (R200Q)VSX2 hiPSC-OVs relative to sibling control hiPSC-OVs. Interestingly, long-term supplementation with FGF9, but not FGF19, partially rescued the mutant retinal phenotype of the (R200Q)VSX2 hiPSC-OV model. However, antagonizing FGF9 in wild-type control hiPSCs did not alter OV development. CONCLUSIONS: Our results show that FGF9 acts in concert with VSX2 to promote NR differentiation in hiPSC-OVs and has potential to be used to manipulate early retinogenesis and mitigate ocular defects caused by functional loss of VSX2 activity. NOTE: Publication of this article is sponsored by the American Ophthalmological Society.


Asunto(s)
Factor 9 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Microftalmía/genética , Epitelio Pigmentado de la Retina/crecimiento & desarrollo , Western Blotting , Diferenciación Celular , Células Cultivadas , Factor 9 de Crecimiento de Fibroblastos/biosíntesis , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , Microftalmía/metabolismo , Microftalmía/patología , Fenotipo , ARN/genética , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/metabolismo
5.
Development ; 146(1)2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30567931

RESUMEN

Numerous protocols have been described for producing neural retina from human pluripotent stem cells (hPSCs), many of which are based on the culture of 3D organoids. Although nearly all such methods yield at least partial segments of retinal structure with a mature appearance, variabilities exist within and between organoids that can change over a protracted time course of differentiation. Adding to this complexity are potential differences in the composition and configuration of retinal organoids when viewed across multiple differentiations and hPSC lines. In an effort to understand better the current capabilities and limitations of these cultures, we generated retinal organoids from 16 hPSC lines and monitored their appearance and structural organization over time by light microscopy, immunocytochemistry, metabolic imaging and electron microscopy. We also employed optical coherence tomography and 3D imaging techniques to assess and compare whole or broad regions of organoids to avoid selection bias. Results from this study led to the development of a practical staging system to reduce inconsistencies in retinal organoid cultures and increase rigor when utilizing them in developmental studies, disease modeling and transplantation.


Asunto(s)
Organoides/citología , Células Madre Pluripotentes/citología , Retina/citología , Diferenciación Celular , Línea Celular , Proliferación Celular , Forma de la Célula , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Humanos , Interneuronas/citología , Interneuronas/metabolismo , Modelos Biológicos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/ultraestructura , Reproducibilidad de los Resultados , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/metabolismo , Sinapsis/metabolismo , Tomografía de Coherencia Óptica
6.
Sci Rep ; 8(1): 2370, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29402929

RESUMEN

Reporter lines generated in human pluripotent stem cells can be highly useful for the analysis of specific cell types and lineages in live cultures. We created the first human rod reporter line using CRISPR/Cas9 genome editing to replace one allele of the Neural Retina Leucine zipper (NRL) gene with an eGFP transgene in the WA09 human embryonic stem cell (hESC) line. After confirming successful targeting, three-dimensional optic vesicle structures were produced to examine reporter specificity and to track rod differentiation in culture. The NRL+/eGFP hESC line robustly and exclusively labeled the entirety of rods throughout differentiation, eventually revealing highly mature structural features. This line provides a valuable tool for studying human rod development and disease and testing therapeutic strategies for retinitis pigmentosa.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/análisis , Diferenciación Celular , Proteínas del Ojo/análisis , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Células Madre Pluripotentes/fisiología , Células Fotorreceptoras Retinianas Bastones/fisiología , Coloración y Etiquetado/métodos , Línea Celular , Edición Génica , Proteínas Fluorescentes Verdes/genética , Humanos , Recombinación Genética
7.
Stem Cells ; 36(3): 313-324, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29230913

RESUMEN

Cell type-specific investigations commonly use gene reporters or single-cell analytical techniques. However, reporter line development is arduous and generally limited to a single gene of interest, while single-cell RNA (scRNA)-sequencing (seq) frequently yields equivocal results that preclude definitive cell identification. To examine gene expression profiles of multiple retinal cell types derived from human pluripotent stem cells (hPSCs), we performed scRNA-seq on optic vesicle (OV)-like structures cultured under cGMP-compatible conditions. However, efforts to apply traditional scRNA-seq analytical methods based on unbiased algorithms were unrevealing. Therefore, we developed a simple, versatile, and universally applicable approach that generates gene expression data akin to those obtained from reporter lines. This method ranks single cells by expression level of a bait gene and searches the transcriptome for genes whose cell-to-cell rank order expression most closely matches that of the bait. Moreover, multiple bait genes can be combined to refine datasets. Using this approach, we provide further evidence for the authenticity of hPSC-derived retinal cell types. Stem Cells 2018;36:313-324.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Retina/citología , Análisis de la Célula Individual/métodos , Perfilación de la Expresión Génica , Humanos , Análisis de Secuencia de ARN/métodos
8.
Stem Cells ; 34(11): 2625-2634, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27301076

RESUMEN

Few gene targets of Visual System Homeobox 2 (VSX2) have been identified despite its broad and critical role in the maintenance of neural retina (NR) fate during early retinogenesis. We performed VSX2 ChIP-seq and ChIP-PCR assays on early stage optic vesicle-like structures (OVs) derived from human iPS cells (hiPSCs), which highlighted WNT pathway genes as direct regulatory targets of VSX2. Examination of early NR patterning in hiPSC-OVs from a patient with a functional null mutation in VSX2 revealed mis-expression and upregulation of WNT pathway components and retinal pigmented epithelium (RPE) markers in comparison to control hiPSC-OVs. Furthermore, pharmacological inhibition of WNT signaling rescued the early mutant phenotype, whereas augmentation of WNT signaling in control hiPSC-OVs phenocopied the mutant. These findings reveal an important role for VSX2 as a regulator of WNT signaling and suggest that VSX2 may act to maintain NR identity at the expense of RPE in part by direct repression of WNT pathway constituents. Stem Cells 2016;34:2625-2634.


Asunto(s)
Tipificación del Cuerpo/genética , Proteínas de Homeodominio/genética , Células Madre Pluripotentes Inducidas/metabolismo , Microftalmía/genética , Epitelio Pigmentado de la Retina/metabolismo , Factores de Transcripción/genética , Proteína Wnt1/genética , Sustitución de Aminoácidos , Benzotiazoles/farmacología , Biomarcadores/metabolismo , Diferenciación Celular , Cuerpos Embrioides/efectos de los fármacos , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/patología , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Microftalmía/metabolismo , Microftalmía/patología , Mutación , Fenotipo , Cultivo Primario de Células , Piridinas/farmacología , Pirimidinas/farmacología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/patología , Factores de Transcripción/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt1/agonistas , Proteína Wnt1/antagonistas & inhibidores , Proteína Wnt1/metabolismo
9.
PLoS One ; 10(8): e0135830, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26292211

RESUMEN

Three dimensional (3D) culture techniques are frequently used for CNS tissue modeling and organoid production, including generation of retina-like tissues. A proposed advantage of these 3D systems is their potential to more closely approximate in vivo cellular microenvironments, which could translate into improved manufacture and/or maintenance of neuronal populations. Visual System Homeobox 2 (VSX2) labels all multipotent retinal progenitor cells (RPCs) and is known to play important roles in retinal development. In contrast, the proneural transcription factor Acheate scute-like 1 (ASCL1) is expressed transiently in a subset of RPCs, but is required for the production of most retinal neurons. Therefore, we asked whether the presence of VSX2 and ASCL1 could gauge neurogenic potential in 3D retinal cultures derived from human prenatal tissue or ES cells (hESCs). Short term prenatal 3D retinal cultures displayed multiple characteristics of human RPCs (hRPCs) found in situ, including robust expression of VSX2. Upon initiation of hRPC differentiation, there was a small increase in co-labeling of VSX2+ cells with ASCL1, along with a modest increase in the number of PKCα+ neurons. However, 3D prenatal retinal cultures lost expression of VSX2 and ASCL1 over time while concurrently becoming refractory to neuronal differentiation. Conversely, 3D optic vesicles derived from hESCs (hESC-OVs) maintained a robust VSX2+ hRPC population that could spontaneously co-express ASCL1 and generate photoreceptors and other retinal neurons for an extended period of time. These results show that VSX2 and ASCL1 can serve as markers for neurogenic potential in cultured hRPCs. Furthermore, unlike hESC-OVs, maintenance of 3D structure does not independently convey an advantage in the culture of prenatal hRPCs, further illustrating differences in the survival and differentiation requirements of hRPCs extracted from native tissue vs. those generated entirely in vitro.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Proteínas de Homeodominio/fisiología , Células-Madre Neurales/fisiología , Retina/citología , Factores de Transcripción/fisiología , Diferenciación Celular/fisiología , Humanos , Imagenología Tridimensional , Neurogénesis/fisiología , Reacción en Cadena de la Polimerasa , Retina/embriología , Retina/fisiología
10.
Hum Mol Genet ; 23(23): 6332-44, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25008112

RESUMEN

Microphthalmia-associated transcription factor (MITF) is a master regulator of pigmented cell survival and differentiation with direct transcriptional links to cell cycle, apoptosis and pigmentation. In mouse, Mitf is expressed early and uniformly in optic vesicle (OV) cells as they evaginate from the developing neural tube, and null Mitf mutations result in microphthalmia and pigmentation defects. However, homozygous mutations in MITF have not been identified in humans; therefore, little is known about its role in human retinogenesis. We used a human embryonic stem cell (hESC) model that recapitulates numerous aspects of retinal development, including OV specification and formation of retinal pigment epithelium (RPE) and neural retina progenitor cells (NRPCs), to investigate the earliest roles of MITF. During hESC differentiation toward a retinal lineage, a subset of MITF isoforms was expressed in a sequence and tissue distribution similar to that observed in mice. In addition, we found that promoters for the MITF-A, -D and -H isoforms were directly targeted by Visual Systems Homeobox 2 (VSX2), a transcription factor involved in patterning the OV toward a NRPC fate. We then manipulated MITF RNA and protein levels at early developmental stages and observed decreased expression of eye field transcription factors, reduced early OV cell proliferation and disrupted RPE maturation. This work provides a foundation for investigating MITF and other highly complex, multi-purposed transcription factors in a dynamic human developmental model system.


Asunto(s)
Células Madre Embrionarias/metabolismo , Factor de Transcripción Asociado a Microftalmía/genética , Células-Madre Neurales/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Células Madre Embrionarias/citología , Técnicas de Inactivación de Genes , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Factor de Transcripción Asociado a Microftalmía/metabolismo , Células-Madre Neurales/citología , Regiones Promotoras Genéticas , Isoformas de Proteínas/metabolismo , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/embriología , Factores de Transcripción/metabolismo
11.
Stem Cells ; 32(6): 1480-92, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24532057

RESUMEN

Human induced pluripotent stem cells (hiPSCs) have been shown to differentiate along the retinal lineage in a manner that mimics normal mammalian development. Under certain culture conditions, hiPSCs form optic vesicle-like structures (OVs), which contain proliferating progenitors capable of yielding all neural retina (NR) cell types over time. Such observations imply conserved roles for regulators of retinogenesis in hiPSC-derived cultures and the developing embryo. However, whether and to what extent this assumption holds true has remained largely uninvestigated. We examined the role of a key NR transcription factor, visual system homeobox 2 (VSX2), using hiPSCs derived from a patient with microphthalmia caused by an R200Q mutation in the VSX2 homeodomain region. No differences were noted between (R200Q)VSX2 and sibling control hiPSCs prior to OV generation. Thereafter, (R200Q)VSX2 hiPSC-OVs displayed a significant growth deficit compared to control hiPSC-OVs, as well as increased production of retinal pigmented epithelium at the expense of NR cell derivatives. Furthermore, (R200Q)VSX2 hiPSC-OVs failed to produce bipolar cells, a distinctive feature previously observed in Vsx2 mutant mice. (R200Q)VSX2 hiPSC-OVs also demonstrated delayed photoreceptor maturation, which could be overcome via exogenous expression of wild-type VSX2 at early stages of retinal differentiation. Finally, RNAseq analysis on isolated hiPSC-OVs implicated key transcription factors and extracellular signaling pathways as potential downstream effectors of VSX2-mediated gene regulation. Our results establish hiPSC-OVs as versatile model systems to study retinal development at stages not previously accessible in humans and support the bona fide nature of hiPSC-OV-derived retinal progeny.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Retina/embriología , Retina/metabolismo , Factores de Transcripción/metabolismo , Adulto , Sustitución de Aminoácidos , Animales , Tipificación del Cuerpo/genética , Diferenciación Celular , Línea Celular , Linaje de la Célula , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Masculino , Ratones , Mutación/genética , Fenotipo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Retina/patología , Células Bipolares de la Retina/metabolismo , Células Bipolares de la Retina/patología , Epitelio Pigmentado de la Retina/embriología , Epitelio Pigmentado de la Retina/patología , Análisis de Secuencia de ARN , Transducción de Señal/genética , Factores de Transcripción/genética , Transcriptoma/genética
12.
Invest Ophthalmol Vis Sci ; 54(10): 6767-78, 2013 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-24030465

RESUMEN

PURPOSE: To determine the effects of serial expansion on the cellular, molecular, and functional properties of human iPS cell (hiPSC)-derived RPE cultures. METHODS: Fibroblasts obtained from four individuals were reprogrammed into hiPSCs and differentiated to RPE cells using previously described methods. Patches of deeply pigmented hiPSC-RPE were dissected, dissociated, and grown in culture until they re-formed pigmented monolayers. Subsequent passages were obtained by repeated dissociation, expansion, and maturation of RPE into pigmented monolayers. Gene and protein expression profiles and morphological and functional characteristics of hiPSC-RPE at different passages were compared with each other and to human fetal RPE (hfRPE). RESULTS: RPE from all four hiPSC lines could be expanded more than 1000-fold when serially passaged as pigmented monolayer cultures. Importantly, expansion of hiPSC-RPE monolayers over the first three passages (P1-P3) resulted in decreased expression of pluripotency and neuroretinal markers and maintenance of characteristic morphological features and gene and protein expression profiles. Furthermore, P1 to P3 hiPSC-RPE monolayers reliably demonstrated functional tight junctions, G-protein-coupled receptor-mediated calcium transients, phagocytosis and degradation of photoreceptor outer segments, and polarized secretion of biomolecules. In contrast, P4 hiPSC-RPE cells failed to form monolayers and possessed altered morphological and functional characteristics and gene expression levels. CONCLUSIONS: Highly differentiated, pigmented hiPSC-RPE monolayers can undergo limited serial expansion while retaining key cytological and functional attributes. However, passaging hiPSC-RPE cultures beyond senescence leads to loss of such features. Our findings support limited, controlled passaging of patient-specific hiPSC-RPE to procure cells needed for in vitro disease modeling, drug screening, and cellular transplantation.


Asunto(s)
Células Madre Embrionarias/citología , Epitelio Pigmentado de la Retina/embriología , Animales , Western Blotting , Bovinos , Diferenciación Celular , Línea Celular , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunohistoquímica , Fagocitosis , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Epitelio Pigmentado de la Retina/metabolismo
13.
Hum Mol Genet ; 22(3): 593-607, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23139242

RESUMEN

Best disease (BD) is an inherited degenerative disease of the human macula that results in progressive and irreversible central vision loss. It is caused by mutations in the retinal pigment epithelium (RPE) gene BESTROPHIN1 (BEST1), which, through mechanism(s) that remain unclear, lead to the accumulation of subretinal fluid and autofluorescent waste products from shed photoreceptor outer segments (POSs). We employed human iPS cell (hiPSC) technology to generate RPE from BD patients and unaffected siblings in order to examine the cellular and molecular processes underlying this disease. Consistent with the clinical phenotype of BD, RPE from mutant hiPSCs displayed disrupted fluid flux and increased accrual of autofluorescent material after long-term POS feeding when compared with hiPSC-RPE from unaffected siblings. On a molecular level, RHODOPSIN degradation after POS feeding was delayed in BD hiPSC-RPE relative to unaffected sibling hiPSC-RPE, directly implicating impaired POS handling in the pathophysiology of the disease. In addition, stimulated calcium responses differed between BD and normal sibling hiPSC-RPE, as did oxidative stress levels after chronic POS feeding. Subcellular localization, fractionation and co-immunoprecipitation experiments in hiPSC-RPE and human prenatal RPE further linked BEST1 to the regulation and release of endoplasmic reticulum calcium stores. Since calcium signaling and oxidative stress are critical regulators of fluid flow and protein degradation, these findings likely contribute to the clinical picture of BD. In a larger context, this report demonstrates the potential to use patient-specific hiPSCs to model and study maculopathies, an important class of blinding disorders in humans.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/fisiopatología , Animales , Bestrofinas , Calcio/metabolismo , Bovinos , Diferenciación Celular , Línea Celular , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Regulación de la Expresión Génica , Homeostasis , Humanos , Inmunohistoquímica , Inmunoprecipitación , Mácula Lútea/patología , Microscopía Electrónica de Transmisión , Estrés Oxidativo , Fagocitosis , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/patología , Distrofia Macular Viteliforme/metabolismo
14.
Invest Ophthalmol Vis Sci ; 53(4): 2007-19, 2012 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-22410558

RESUMEN

PURPOSE: We sought to determine if human induced pluripotent stem cells (iPSCs) derived from blood could produce optic vesicle-like structures (OVs) with the capacity to stratify and express markers of intercellular communication. METHODS: Activated T-lymphocytes from a routine peripheral blood sample were reprogrammed by retroviral transduction to iPSCs. The T-lymphocyte-derived iPSCs (TiPSCs) were characterized for pluripotency and differentiated to OVs using our previously published protocol. TiPSC-OVs were then manually isolated, pooled, and cultured en masse to more mature stages of retinogenesis. Throughout this stepwise differentiation process, changes in anterior neural, retinal, and synaptic marker expression were monitored by PCR, immunocytochemistry, and/or flow cytometry. RESULTS: TiPSCs generated abundant OVs, which contained a near homogeneous population of proliferating neuroretinal progenitor cells (NRPCs). These NRPCs differentiated into multiple neuroretinal cell types, similar to OV cultures from human embryonic stem cells and fibroblast-derived iPSCs. In addition, portions of some TiPSC-OVs maintained their distinctive neuroepithelial appearance and spontaneously formed primitive laminae, reminiscent of the developing retina. Retinal progeny from TiPSC-OV cultures expressed numerous genes and proteins critical for synaptogenesis and gap junction formation, concomitant with the emergence of glia and the upregulation of thrombospondins in culture. CONCLUSIONS: We demonstrate for the first time that human blood-derived iPSCs can generate retinal cell types, providing a highly convenient donor cell source for iPSC-based retinal studies. We also show that cultured TiPSC-OVs have the capacity to self-assemble into rudimentary neuroretinal structures and express markers indicative of chemical and electrical synapses.


Asunto(s)
Células Madre Pluripotentes Inducidas/fisiología , Morfogénesis , Retina/crecimiento & desarrollo , Sinapsis/fisiología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/citología , Retina/citología , Retina/metabolismo
15.
Stem Cells ; 29(8): 1206-18, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21678528

RESUMEN

Differentiation methods for human induced pluripotent stem cells (hiPSCs) typically yield progeny from multiple tissue lineages, limiting their use for drug testing and autologous cell transplantation. In particular, early retina and forebrain derivatives often intermingle in pluripotent stem cell cultures, owing to their shared ancestry and tightly coupled development. Here, we demonstrate that three-dimensional populations of retinal progenitor cells (RPCs) can be isolated from early forebrain populations in both human embryonic stem cell and hiPSC cultures, providing a valuable tool for developmental, functional, and translational studies. Using our established protocol, we identified a transient population of optic vesicle (OV)-like structures that arose during a time period appropriate for normal human retinogenesis. These structures were independently cultured and analyzed to confirm their multipotent RPC status and capacity to produce physiologically responsive retinal cell types, including photoreceptors and retinal pigment epithelium (RPE). We then applied this method to hiPSCs derived from a patient with gyrate atrophy, a retinal degenerative disease affecting the RPE. RPE generated from these hiPSCs exhibited a disease-specific functional defect that could be corrected either by pharmacological means or following targeted gene repair. The production of OV-like populations from human pluripotent stem cells should facilitate the study of human retinal development and disease and advance the use of hiPSCs in personalized medicine.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Células Madre Pluripotentes/fisiología , Enfermedades de la Retina/terapia , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Expresión Génica , Terapia Genética , Atrofia Girata/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Potenciales de la Membrana , Técnicas de Placa-Clamp , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Células Fotorreceptoras/fisiología , Medicina de Precisión , Prosencéfalo/embriología , Retina/embriología , Retina/patología , Epitelio Pigmentado de la Retina/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Invest Ophthalmol Vis Sci ; 51(4): 2269-76, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19959642

RESUMEN

Purpose. Usher's syndrome is a combined deafness and blindness disorder caused by mutations in several genes with functions in both the retina and the ear. Here the authors studied morphologic and functional changes in an animal model, the Ush2a mouse, and explored whether transplantation of forebrain-derived progenitor cells might affect the progress of morphologic and functional deterioration. Methods. Ush2a mice were tested at postnatal days (P) 70 to P727 using an optomotor test, which provides a repeatable method of estimating rodent visual acuity and contrast sensitivity. A group of mice that received grafts of forebrain-derived progenitor cells at P80 was tested for up to 10 weeks after grafting. At the end of testing, animals were killed, and eyes were processed for histology. Results. The optomotor test showed that both acuity and contrast sensitivity deteriorated over time; contrast sensitivity showed a deficit even at P70. By contrast, photoreceptor loss was only evident later than 1 year of age, though changes in the intracellular distribution of red/green cone opsin were observed as early as P80. Mice that received transplanted cells performed significantly better than control mice and no longer demonstrated abnormal distribution of red/green opsin where the donor cells were distributed. Conclusions. This study showed that vision impairment was detected well before significant photoreceptor loss and was correlated with abnormal distribution of a cone pigment. Cell transplantation prevented functional deterioration for at least 10 weeks and reversed the mislocalization of cone pigment.


Asunto(s)
Modelos Animales de Enfermedad , Trasplante de Células Madre , Trasplante Heterólogo , Síndromes de Usher/fisiopatología , Síndromes de Usher/cirugía , Animales , Trasplante de Células , Sensibilidad de Contraste/fisiología , Proteínas de la Matriz Extracelular/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Opsinas/metabolismo , Prosencéfalo/citología , Retina/fisiopatología , Células Fotorreceptoras Retinianas Conos/metabolismo , Degeneración Retiniana/fisiopatología , Degeneración Retiniana/cirugía , Umbral Sensorial , Células Madre/fisiología , Síndromes de Usher/genética , Trastornos de la Visión/genética , Trastornos de la Visión/fisiopatología , Trastornos de la Visión/cirugía , Agudeza Visual/fisiología
17.
Proc Natl Acad Sci U S A ; 106(39): 16698-703, 2009 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-19706890

RESUMEN

Human pluripotent stem cells have the potential to provide comprehensive model systems for the earliest stages of human ontogenesis. To serve in this capacity, these cells must undergo a targeted, stepwise differentiation process that follows a normal developmental timeline. Here we demonstrate the ability of both human embryonic stem cells (hESCs) and induced pluripotent stem (iPS) cells to meet these requirements for human retinogenesis. Upon differentiation, hESCs initially yielded a highly enriched population of early eye field cells. Thereafter, a subset of cells acquired features of advancing retinal differentiation in a sequence and time course that mimicked in vivo human retinal development. Application of this culture method to a human iPS cell line also generated retina-specific cell types at comparable times in vitro. Lastly, altering endogenous signaling during differentiation affected lineage-specific gene expression in a manner consistent with established mechanisms of early neural and retinal cell fate determination. These findings should aid in the investigation of the molecular events governing retinal specification from human pluripotent stem cells.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Pluripotentes/citología , Retina/crecimiento & desarrollo , Diferenciación Celular , Células Madre Embrionarias/metabolismo , Humanos , Inmunohistoquímica , Modelos Biológicos , Fenotipo , Células Madre Pluripotentes/metabolismo , Retina/embriología
18.
Stem Cells ; 26(12): 3182-93, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18802035

RESUMEN

During development of the central nervous system, stem and progenitor cell proliferation and differentiation are controlled by complex inter- and intracellular interactions that orchestrate the precise spatiotemporal production of particular cell types. Within the embryonic retina, progenitor cells are located adjacent to the retinal pigment epithelium (RPE), which differentiates prior to the neurosensory retina and has the capacity to secrete a multitude of growth factors. We found that secreted proteinaceous factors in human prenatal RPE conditioned medium (RPE CM) prolonged and enhanced the growth of human prenatal retinal neurospheres. The growth-promoting activity of RPE CM was mitogen-dependent and associated with an acute increase in transcription factor phosphorylation. Expanded populations of RPE CM-treated retinal neurospheres expressed numerous neurodevelopmental and eye specification genes and markers characteristic of neural and retinal progenitor cells, but gradually lost the potential to generate neurons upon differentiation. Misexpression of Mash1 restored the neurogenic potential of long-term cultures, yielding neurons with phenotypic characteristics of multiple inner retinal cell types. Thus, a novel combination of extrinsic and intrinsic factors was required to promote both progenitor cell proliferation and neuronal multipotency in human retinal neurosphere cultures. These results support a pro-proliferative and antiapoptotic role for RPE in human retinal development, reveal potential limitations of human retinal progenitor culture systems, and suggest a means for overcoming cell fate restriction in vitro.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Neuronas/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Apoptosis , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Medios de Cultivo Condicionados/metabolismo , Humanos , Fenotipo , Fosforilación , Retina/citología , Células Madre/citología , Factores de Transcripción/metabolismo
19.
Stem Cells Dev ; 17(1): 107-17, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18225979

RESUMEN

Human stem and progenitor cells offer an innovative way to study early events in development. An exciting new opportunity for these cells is their application to study the underlying developmental consequences of genetic diseases. Because many diseases, ranging from leukemias to developmental disorders, are caused by single-gene defects, stem and progenitor cells that carry disease-causing genetic mutations are invaluable in understanding and treating disease. We have characterized human neural progenitor (hNPCs) cells that carry a single-gene defect that leads to the neurodevelopmental disorder Fragile X syndrome (FX). A loss-of-function mutation in the FMR1 gene leads to subtle changes in neural development and subsequent mental impairment characteristic of FX. hNPCs were isolated from fetal cortex carrying the FMR1 mutation to determine whether aberrations occur in their proliferation and differentiation. As expected, FX hNPCs have reduced expression of the FMR1 gene product Fragile X mental retardation protein (FMRP), and this decrease is maintained in culture and following differentiation. In contrast to a previously published report, the proliferation of FX hNPCs and their differentiation into neurons is not different from unaffected controls. Although the early development of FX hNPCs is essentially normal, microarray analysis reveals novel changes in the expression of signal transduction genes in FX hNPCs. Therefore, hNPCs have intrinsic characteristics that can be investigated to further our understanding and potential treatment of developmental disorders such as FX.


Asunto(s)
Corteza Cerebral/patología , Células Madre Fetales/patología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/patología , Mutación , Neuronas/citología , Diferenciación Celular , Proliferación Celular , Corteza Cerebral/embriología , Humanos
20.
J Neurosci Methods ; 163(2): 338-49, 2007 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-17397931

RESUMEN

Human neural progenitor cells (hNPC) hold great potential as an ex vivo system for delivery of therapeutic proteins to the central nervous system. When cultured as aggregates, termed neurospheres, hNPC are capable of significant in vitro expansion. In the current study, we present a robust method for lentiviral vector-mediated gene delivery into hNPC that maintains the differentiation and proliferative properties of neurosphere cultures while minimizing the amount of viral vector used and controlling the number of insertion sites per population. This method results in long-term, stable expression even after differentiation of the hNPC to neurons and astrocytes and allows for generation of equivalent transgenic populations of hNPC. In addition, the in vitro analysis presented predicts the behavior of transgenic lines in vivo when transplanted into a rodent model of Parkinson's disease. The methods presented provide a powerful tool for assessing the impact of factors such as promoter systems or different transgenes on the therapeutic utility of these cells.


Asunto(s)
Técnicas de Transferencia de Gen/normas , Terapia Genética/métodos , Vectores Genéticos/genética , Lentivirus/genética , Células Madre/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/virología , Biomarcadores/metabolismo , Trasplante de Tejido Encefálico/métodos , Diferenciación Celular/genética , Proliferación Celular , Células Cultivadas , Feto , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Humanos , Neuronas/metabolismo , Neuronas/virología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/terapia , Regiones Promotoras Genéticas/genética , Ratas , Ratas Endogámicas Lew , Esferoides Celulares/citología , Esferoides Celulares/fisiología , Esferoides Celulares/virología , Células Madre/virología , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...