Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biomed Opt Express ; 13(6): 3476-3492, 2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35781966

RESUMO

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.

3.
Cell Stem Cell ; 29(3): 460-471.e3, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35104442

RESUMO

High-definition vision in humans and nonhuman primates is initiated by cone photoreceptors located within a specialized region of the retina called the fovea. Foveal cone death is the ultimate cause of central blindness in numerous retinal dystrophies, including macular degenerative diseases. 3D retinal organoids (ROs) derived from human pluripotent stem cells (hPSCs) hold tremendous promise to model and treat such diseases. To achieve this goal, RO cones should elicit robust and intrinsic light-evoked electrical responses (i.e., phototransduction) akin to adult foveal cones, which has not yet been demonstrated. Here, we show strong, graded, repetitive, and wavelength-specific light-evoked responses from RO cones. The photoresponses and membrane physiology of a significant fraction of these lab-generated cones are comparable with those of intact ex vivo primate fovea. These results greatly increase confidence in ROs as potential sources of functional human cones for cell replacement therapies, drug testing, and in vitro models of retinal dystrophies.


Assuntos
Células-Tronco Pluripotentes , Distrofias Retinianas , Animais , Humanos , Organoides , Primatas , Espécies Reativas de Oxigênio , Retina , Células Fotorreceptoras Retinianas Cones
4.
Commun Biol ; 3(1): 82, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-32081919

RESUMO

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.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Proteínas do Olho/genética , Células-Tronco Pluripotentes Induzidas/fisiologia , Organoides/patologia , Retina/patologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Fatores de Transcrição de Zíper de Leucina Básica/deficiência , Estudos de Casos e Controles , Diferenciação Celular/genética , Células Cultivadas , Reprogramação Celular/fisiologia , Oftalmopatias Hereditárias/genética , Oftalmopatias Hereditárias/patologia , Feto/patologia , Perfilação da Expressão Gênica , Humanos , Regeneração Nervosa/genética , Neurogênese/genética , Organoides/fisiologia , Cultura Primária de Células/métodos , Retina/fisiologia , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Retinose Pigmentar/genética , Retinose Pigmentar/patologia , Transcriptoma , Transtornos da Visão/genética , Transtornos da Visão/patologia
5.
Hum Mol Genet ; 29(6): 967-979, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32011687

RESUMO

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.


Assuntos
Marcadores Genéticos , Variação Genética , Genoma Humano , RNA-Seq/métodos , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Sequenciamento Completo do Genoma/métodos , Criança , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Linhagem , Sequenciamento do Exoma
6.
Am J Ophthalmol ; 206: 113-131, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31078532

RESUMO

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.


Assuntos
Fator 9 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Pluripotentes Induzidas/citologia , Microftalmia/genética , Epitélio Pigmentado da Retina/crescimento & desenvolvimento , Western Blotting , Diferenciação Celular , Células Cultivadas , Fator 9 de Crescimento de Fibroblastos/biossíntese , Humanos , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas/metabolismo , Microftalmia/metabolismo , Microftalmia/patologia , Fenótipo , RNA/genética , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo
7.
Am J Hum Genet ; 104(2): 310-318, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30686507

RESUMO

Pathogenic variants of the KCNJ13 gene are known to cause Leber congenital amaurosis (LCA16), an inherited pediatric blindness. KCNJ13 encodes the Kir7.1 subunit that acts as a tetrameric, inwardly rectifying potassium ion channel in the retinal pigment epithelium (RPE) to maintain ionic homeostasis and allow photoreceptors to encode visual information. We sought to determine whether genetic approaches might be effective in treating blindness arising from pathogenic variants in KCNJ13. We derived human induced pluripotent stem cell (hiPSC)-RPE cells from an individual carrying a homozygous c.158G>A (p.Trp53∗) pathogenic variant of KCNJ13. We performed biochemical and electrophysiology assays to confirm Kir7.1 function. We tested both small-molecule readthrough drug and gene-therapy approaches for this "disease-in-a-dish" approach. We found that the LCA16 hiPSC-RPE cells had normal morphology but did not express a functional Kir7.1 channel and were unable to demonstrate normal physiology. After readthrough drug treatment, the LCA16 hiPSC cells were hyperpolarized by 30 mV, and the Kir7.1 current was restored. Similarly, we rescued Kir7.1 channel function after lentiviral gene delivery to the hiPSC-RPE cells. In both approaches, Kir7.1 was expressed normally, and there was restoration of membrane potential and the Kir7.1 current. Loss-of-function variants of Kir7.1 are one cause of LCA. Using either readthrough therapy or gene augmentation, we rescued Kir7.1 channel function in iPSC-RPE cells derived from an affected individual. This supports the development of precision-medicine approaches for the treatment of clinical LCA16.


Assuntos
Cegueira/congênito , Canalopatias/genética , Terapia Genética/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Amaurose Congênita de Leber/genética , Modelos Biológicos , Canais de Potássio Corretores do Fluxo de Internalização/genética , Epitélio Pigmentado da Retina/patologia , Sequência de Bases , Cegueira/genética , Cegueira/patologia , Canalopatias/patologia , Criança , Humanos , Amaurose Congênita de Leber/patologia , Epitélio Pigmentado da Retina/metabolismo
8.
Development ; 146(1)2019 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-30567931

RESUMO

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.


Assuntos
Organoides/citologia , Células-Tronco Pluripotentes/citologia , Retina/citologia , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Forma Celular , Células Ependimogliais/citologia , Células Ependimogliais/metabolismo , Humanos , Interneurônios/citologia , Interneurônios/metabolismo , Modelos Biológicos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/ultraestrutura , Reprodutibilidade dos Testes , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/metabolismo , Sinapses/metabolismo , Tomografia de Coerência Óptica
9.
Sci Rep ; 8(1): 2370, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29402929

RESUMO

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.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/análise , Diferenciação Celular , Proteínas do Olho/análise , Genes Reporter , Proteínas de Fluorescência Verde/análise , Células-Tronco Pluripotentes/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Coloração e Rotulagem/métodos , Linhagem Celular , Edição de Genes , Proteínas de Fluorescência Verde/genética , Humanos , Recombinação Genética
10.
Stem Cells ; 36(3): 313-324, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29230913

RESUMO

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.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala/métodos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Retina/citologia , Análise de Célula Única/métodos , Perfilação da Expressão Gênica , Humanos , Análise de Sequência de RNA/métodos
11.
Stem Cells ; 34(11): 2625-2634, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27301076

RESUMO

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.


Assuntos
Padronização Corporal/genética , Proteínas de Homeodomínio/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Microftalmia/genética , Epitélio Pigmentado da Retina/metabolismo , Fatores de Transcrição/genética , Proteína Wnt1/genética , Substituição de Aminoácidos , Benzotiazóis/farmacologia , Biomarcadores/metabolismo , Diferenciação Celular , Corpos Embrioides/efeitos dos fármacos , Corpos Embrioides/metabolismo , Corpos Embrioides/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/patologia , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Microftalmia/metabolismo , Microftalmia/patologia , Mutação , Fenótipo , Cultura Primária de Células , Piridinas/farmacologia , Pirimidinas/farmacologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/patologia , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt1/agonistas , Proteína Wnt1/antagonistas & inibidores , Proteína Wnt1/metabolismo
12.
PLoS One ; 10(8): e0135830, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26292211

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas de Homeodomínio/fisiologia , Células-Tronco Neurais/fisiologia , Retina/citologia , Fatores de Transcrição/fisiologia , Diferenciação Celular/fisiologia , Humanos , Imageamento Tridimensional , Neurogênese/fisiologia , Reação em Cadeia da Polimerase , Retina/embriologia , Retina/fisiologia
13.
Hum Mol Genet ; 23(23): 6332-44, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25008112

RESUMO

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.


Assuntos
Células-Tronco Embrionárias/metabolismo , Fator de Transcrição Associado à Microftalmia/genética , Células-Tronco Neurais/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Células-Tronco Embrionárias/citologia , Técnicas de Inativação de Genes , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Fator de Transcrição Associado à Microftalmia/metabolismo , Células-Tronco Neurais/citologia , Regiões Promotoras Genéticas , Isoformas de Proteínas/metabolismo , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/embriologia , Fatores de Transcrição/metabolismo
14.
Stem Cells ; 32(6): 1480-92, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24532057

RESUMO

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.


Assuntos
Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Retina/embriologia , Retina/metabolismo , Fatores de Transcrição/metabolismo , Adulto , Substituição de Aminoácidos , Animais , Padronização Corporal/genética , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Células HEK293 , Proteínas de Homeodomínio/genética , Humanos , Masculino , Camundongos , Mutação/genética , Fenótipo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patologia , Retina/patologia , Células Bipolares da Retina/metabolismo , Células Bipolares da Retina/patologia , Epitélio Pigmentado da Retina/embriologia , Epitélio Pigmentado da Retina/patologia , Análise de Sequência de RNA , Transdução de Sinais/genética , Fatores de Transcrição/genética , Transcriptoma/genética
15.
Invest Ophthalmol Vis Sci ; 54(10): 6767-78, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24030465

RESUMO

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.


Assuntos
Células-Tronco Embrionárias/citologia , Epitélio Pigmentado da Retina/embriologia , Animais , Western Blotting , Bovinos , Diferenciação Celular , Linhagem Celular , Proteínas do Olho/biossíntese , Proteínas do Olho/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imuno-Histoquímica , Fagocitose , RNA/genética , Reação em Cadeia da Polimerase em Tempo Real , Epitélio Pigmentado da Retina/metabolismo
16.
Hum Mol Genet ; 22(3): 593-607, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23139242

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/fisiopatologia , Animais , Bestrofinas , Cálcio/metabolismo , Bovinos , Diferenciação Celular , Linhagem Celular , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica , Homeostase , Humanos , Imuno-Histoquímica , Imunoprecipitação , Macula Lutea/patologia , Microscopia Eletrônica de Transmissão , Estresse Oxidativo , Fagocitose , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/patologia , Distrofia Macular Viteliforme/metabolismo
17.
Invest Ophthalmol Vis Sci ; 53(4): 2007-19, 2012 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-22410558

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas/fisiologia , Morfogênese , Retina/crescimento & desenvolvimento , Sinapses/fisiologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Retina/citologia , Retina/metabolismo
18.
Stem Cells ; 29(8): 1206-18, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21678528

RESUMO

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.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Células-Tronco Pluripotentes/fisiologia , Doenças Retinianas/terapia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Expressão Gênica , Terapia Genética , Atrofia Girata/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Potenciais da Membrana , Técnicas de Patch-Clamp , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patologia , Células Fotorreceptoras/fisiologia , Medicina de Precisão , Prosencéfalo/embriologia , Retina/embriologia , Retina/patologia , Epitélio Pigmentado da Retina/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
19.
Invest Ophthalmol Vis Sci ; 51(4): 2269-76, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19959642

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Transplante de Células-Tronco , Transplante Heterólogo , Síndromes de Usher/fisiopatologia , Síndromes de Usher/cirurgia , Animais , Transplante de Células , Sensibilidades de Contraste/fisiologia , Proteínas da Matriz Extracelular/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Opsinas/metabolismo , Prosencéfalo/citologia , Retina/fisiopatologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Degeneração Retiniana/fisiopatologia , Degeneração Retiniana/cirurgia , Limiar Sensorial , Células-Tronco/fisiologia , Síndromes de Usher/genética , Transtornos da Visão/genética , Transtornos da Visão/fisiopatologia , Transtornos da Visão/cirurgia , Acuidade Visual/fisiologia
20.
Proc Natl Acad Sci U S A ; 106(39): 16698-703, 2009 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-19706890

RESUMO

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.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes/citologia , Retina/crescimento & desenvolvimento , Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Humanos , Imuno-Histoquímica , Modelos Biológicos , Fenótipo , Células-Tronco Pluripotentes/metabolismo , Retina/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA