Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Development ; 147(12)2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32467236

RESUMEN

The zebrafish retina grows for a lifetime. Whether embryonic and postembryonic retinogenesis conform to the same developmental program is an outstanding question that remains under debate. Using single-cell RNA sequencing of ∼20,000 cells of the developing zebrafish retina at four different stages, we identified seven distinct developmental states. Each state explicitly expresses a gene set. Disruption of individual state-specific marker genes results in various defects ranging from small eyes to the loss of distinct retinal cell types. Using a similar approach, we further characterized the developmental states of postembryonic retinal stem cells (RSCs) and their progeny in the ciliary marginal zone. Expression pattern analysis of state-specific marker genes showed that the developmental states of postembryonic RSCs largely recapitulated those of their embryonic counterparts, except for some differences in rod photoreceptor genesis. Thus, our findings reveal the unifying developmental program used by the embryonic and postembryonic retinogenesis in zebrafish.


Asunto(s)
Embrión no Mamífero/metabolismo , Neurogénesis/genética , Retina/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente/crecimiento & desarrollo , Animales Modificados Genéticamente/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario , Proteínas de Unión a Ácidos Grasos/deficiencia , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Retina/citología , Retina/crecimiento & desarrollo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
2.
Cell Tissue Res ; 392(3): 689-704, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36802303

RESUMEN

Retinal degenerative diseases such as glaucoma, retinitis pigmentosa, and age-related macular degeneration pose serious threats to human visual health due to lack of effective therapeutic approaches. In recent years, the transplantation of retinal progenitor cells (RPCs) has shown increasing promise in the treatment of these diseases; however, the application of RPC transplantation is limited by both their poor proliferation and their differentiation capabilities. Previous studies have shown that microRNAs (miRNA) act as essential mediators in the fate determination of stem/progenitor cells. In this study, we hypothesized that miR-124-3p plays a regulatory role in the fate of RPC determination by targeting Septin10 (SEPT10) in vitro. We observed that the overexpression of miR124-3p downregulates SEPT10 expression in RPCs, leading to reduced RPC proliferation and increased differentiation, specifically towards both neurons and ganglion cells. Conversely, antisense knockdown of miR-124-3p was shown to boost SEPT10 expression, enhance RPC proliferation, and attenuate differentiation. Moreover, overexpression of SEPT10 rescued miR-124-3p-caused proliferation deficiency while weakening the enhancement of miR-124-3p-induced-RPC differentiation. Results from this study show that miR-124-3p regulates RPC proliferation and differentiation by targeting SEPT10. Furthermore, our findings enable a more comprehensive understanding into the mechanisms of proliferation and differentiation of RPC fate determination. Ultimately, this study may be useful for helping researchers and clinicians to develop more promising and effective approaches to optimize the use of RPCs in treating retinal degeneration diseases.


Asunto(s)
MicroARNs , Degeneración Retiniana , Humanos , Proliferación Celular/genética , Células Cultivadas , Células Madre , Diferenciación Celular/genética , MicroARNs/genética , MicroARNs/metabolismo
3.
Stem Cells ; 40(6): 592-604, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35263762

RESUMEN

The development of the vertebrate retina relies on complex regulatory mechanisms to achieve its characteristic layered morphology containing multiple neuronal cell types. While connexin 43 (CX43) is not expressed by mature retinal neurons, mutations in its gene GJA1 are associated with microphthalmia and low vision in patients. To delineate how lack of CX43 affects retinal development, GJA1 was disrupted in human induced pluripotent stem cells (hiPSCs) (GJA1-/-) using CRISPR/Cas9 editing, and these were subsequently differentiated into retinal organoids. GJA1-/- hiPSCs do not display defects in self-renewal and pluripotency, but the resulting organoids are smaller with a thinner neural retina and decreased abundance of many retinal cell types. CX43-deficient organoids express lower levels of the neural marker PAX6 and the retinal progenitor cell (RPC) markers PAX6, SIX3, and SIX6. Conversely, expression of the early neuroectoderm markers SOX1 and SOX2 remains high in GJA1-/- organoids throughout their development. The lack of CX43 results in an increased population of CHX10-positive RPCs that are smaller, disorganized, do not become polarized, and possess a limited ability to commit to retinal fate specification. Our data indicate that lack of CX43 causes a developmental arrest in RPCs that subsequently leads to pan-retinal defects and stunted ocular growth.


Asunto(s)
Células Madre Pluripotentes Inducidas , Organoides , Diferenciación Celular/genética , Conexina 43/genética , Conexina 43/metabolismo , Humanos , Retina
4.
J Cell Mol Med ; 26(11): 3254-3268, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35481949

RESUMEN

Despite notable efforts and significant therapeutical advances, age-related macular degeneration remains the single most common reason for vision loss. Retinal progenitor cells (RPCs) are considered promising candidates for cellular treatments that repair and restore vision. In this allogenic study, the phenotypic profile of pig and human RPCs derived using similar manufacturing processes is compared. The long-term (12-week) survival of green fluorescent protein-pig retinal progenitor cells GFP-pRPC after subretinal transplantation into normal miniature pig (mini-pig) retina is investigated. Human eyes are both anatomically and physiologically mimicked by pig eyes, so the pig is an ideal model to show an equivalent way of delivering cells, immunological response and dosage. The phenotypic equivalency of porcine and clinically intended human RPCs was established. Thirty-nine mini-pigs are used in this study, and vehicle-injected eyes and non-injected eyes serve as controls. Six groups are given different dosages of pRPCs, and the cells are found to survive well in all groups. At 12 weeks, strong evidence of integration is indicated by the location of the grafted cells within the neuro-retina, extension of processes to the plexiform layers and expression of key retinal markers such as recoverin, rhodopsin and synaptophysin. No immunosuppression is used, and no immune response is found in any of the groups. No pRPC-related histopathology findings are reported in the major organs investigated. An initial dose of 250 k cells in 100 µl of buffer is established as an appropriate initial dose for future human clinical trials.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Retina , Animales , Diferenciación Celular/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Retina/metabolismo , Trasplante de Células Madre , Porcinos , Porcinos Enanos
5.
Dev Biol ; 478: 41-58, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34146533

RESUMEN

Recent advances in high throughput single-cell RNA sequencing (scRNA-seq) technology have enabled the simultaneous transcriptomic profiling of thousands of individual cells in a single experiment. To investigate the intrinsic process of retinal development, researchers have leveraged this technology to quantify gene expression in retinal cells across development, in multiple species, and from numerous important models of human disease. In this review, we summarize recent applications of scRNA-seq and discuss how these datasets have complemented and advanced our understanding of retinal progenitor cell competence, cell fate specification, and differentiation. Finally, we also highlight the outstanding questions in the field that advances in single-cell data generation and analysis will soon be able to answer.


Asunto(s)
Células Madre Multipotentes/citología , RNA-Seq , Retina/crecimiento & desarrollo , Neuronas Retinianas/citología , Análisis de la Célula Individual , Animales , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Multipotentes/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Retina/citología , Retina/embriología , Enfermedades de la Retina/genética , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Neuronas Retinianas/metabolismo , Transcriptoma
6.
Dev Biol ; 453(2): 141-154, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31145883

RESUMEN

The Crb1 and 2 (Crumbs homolog 1 & 2) genes encode large, single-pass transmembrane proteins essential for the apicobasal polarity and adhesion of epithelial cells. Crb1 mutations cause degenerative retinal diseases in humans, including Leber congenital amaurosis type 8 (LCA8) and retinitis pigmentosa type 12 (RP12). In LCA8, impaired photoreceptor development and/or survival is thought to cause blindness during early infancy, whereas, in RP12, progressive photoreceptor degeneration damages peripheral vision later in life. There are multiple animal models of RP12 pathology, but no experimental model of LCA8 recapitulates the full spectrum of its pathological features. To generate a mouse model of LCA8 and identify the functions of Crb1/2 in developing ocular tissues, we used an mRx-Cre driver to generate allelic combinations that enabled conditional gene ablation from the optic vesicle stage. In this series only Crb1/2 double knockout (dKO) mice exhibited characteristics of human LCA8 disease: locally thickened retina with spots devoid of cells, aberrant positioning of retinal cells, severely disrupted lamination, and depigmented retinal-pigmented epithelium. Retinal defects antedated E12.5, which is far earlier than the stage at which photoreceptor cells mainly differentiate. Most remarkably, Crb1/Crb2 dKO showed a severely attenuated electroretinogram at the eye opening stage. These results suggest that human LCA8 can be modeled in the mouse by simultaneously ablating Crb1/2 from the beginning of eye development. Importantly, they also indicate that LCA8 is caused by malfunction of retinal progenitor cells during early ocular development rather than by defective photoreceptor-Muller glial interaction, a mechanism proposed for RP12.


Asunto(s)
Ojo/metabolismo , Ojo/patología , Eliminación de Gen , Amaurosis Congénita de Leber/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Adulto , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Humanos , Amaurosis Congénita de Leber/patología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Especificidad de Órganos , Células Fotorreceptoras de Vertebrados/metabolismo , Pigmentación , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología
7.
BMC Evol Biol ; 20(1): 129, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33008334

RESUMEN

The primordial eye field of the vertebrate embryo is a single entity of retinal progenitor cells spanning the anterior neural plate before bifurcating to form bilateral optic vesicles. Here we review fate mapping data from zebrafish suggesting that prior to evagination of the optic vesicles the eye field may undergo a Maypole-plait migration of progenitor cells through the midline influenced by the anteriorly subducting diencephalon. Such an enigmatic translocation of scaffolding progenitors could have evolutionary significance if pointing, by way of homology, to an ancient mechanism for transition of the single eye field in chordates to contralateral eye fields in vertebrates.


Asunto(s)
Movimiento Celular , Ojo/embriología , Regulación del Desarrollo de la Expresión Génica , Retina/embriología , Pez Cebra/embriología , Animales , Desarrollo Embrionario , Células Madre/citología
8.
Biochem Cell Biol ; 98(1): 50-60, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-30673303

RESUMEN

During a developmental period that extends postnatally in the mouse, proliferating multipotent retinal progenitor cells produce one of 7 major cell types (rod, cone, bipolar, horizontal, amacrine, ganglion, and Müller glial cells) as they exit the cell cycle in consecutive waves. Cell production in the retina is tightly regulated by intrinsic, extrinsic, spatial, and temporal cues, and is coupled to the timing of cell cycle exit. Arsenic-resistance protein 2 (ARS2, also known as SRRT) is a component of the nuclear cap-binding complex involved in RNA Polymerase II transcription, and is required for cell cycle progression. We show that postnatal retinal progenitor cells (RPCs) require ARS2 for proper progression through S phase, and ARS2 disruption leads to early exit from the cell cycle. Furthermore, we observe an increase in the proportion of cells expressing a rod photoreceptor marker, and a loss of Müller glia marker expression, indicating a role for ARS2 in regulating cell fate specification or differentiation. Knockdown of Flice Associated Huge protein (FLASH), which interacts with ARS2 and is required for cell cycle progression and 3'-end processing of replication-dependent histone transcripts, phenocopies ARS2 knockdown. These data implicate ARS2-FLASH-mediated histone mRNA processing in regulating RPC cell cycle kinetics and neuroglial cell fate specification during postnatal retinal development.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Retina/citología , Retina/metabolismo , Fase S , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Ratones , Fenotipo , Factores de Transcripción/genética
9.
Biochem Biophys Res Commun ; 523(2): 493-499, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-31894018

RESUMEN

MicroRNAs have a vital effect on the differentiation of many types of progenitor cells. Recent studies have suggested that miR-17 plays an important role in the differentiation process of brain neural progenitor cells (NPC). Nevertheless, its detailed functions in regulating retinal progenitor cells (RPC) remain unclear. In our study, overexpression and knockdown of miR-17 were performed by transfecting RPC with mimics and inhibitors, respectively. Next, we investigated the role of miR-17 in RPC proliferation and differentiation by the following experiments: qPCR, CCK8, Edu staining, immunostaining and Western blot. The results revealed that miR-17 inhibited RPC proliferation but enhanced differentiation. Furthermore, according to a web-based database analysis, we identified charged multivesicular body protein 1A (CHMP1A) as a target gene. A dual luciferase reporter system showed that miR-17 specifically binds to the CHMP1A 3' untranslated region (UTR). Next, our data showed upregulation of miR-17 decreased CHMP1A protein level, causing reduced proliferation and enhanced differentiation of RPC. Downregulation of miR-17 led to enhanced CHMP1A protein expression, increased RPC proliferation and decreased differentiation. Taken together, our data provide a proven pathway by which miR-17 regulates RPC proliferation and differentiation by targeting CHMP1A.


Asunto(s)
Diferenciación Celular/genética , MicroARNs/genética , Retina/citología , Proteínas de Transporte Vesicular/genética , Regiones no Traducidas 3' , Animales , Proliferación Celular/genética , Células Cultivadas , Ratones Endogámicos C57BL , Células Madre/citología , Células Madre/fisiología
10.
Mol Biol Rep ; 47(3): 1613-1623, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31950326

RESUMEN

Using stem and progenitor cells to treat retinal disorders holds great promise. Using defined culture conditions to maintain the desires phenotype is of utmost clinical importance. We cultured human retinal progenitor cells (hRPCs) in different conditions: such as normoxia (20% oxygen), and hypoxia (5% oxygen) with and without knock-out serum replacement (KOSR) to evaluate its effect on these cells. KOSR is known nutrient supplement often used to replace bovine serum for culturing embryonic or pluripotent stem cells, especially those destined for clinical applications. The purpose of this study was to identify the impact of different environmental and chemical cues to determine if this alters the fate of these cells. Our results indicate that cells cultured with or without KOSR do not show significant differences in viability, but that the oxygen tension can significantly change their viability (higher in hypoxia than normoxia). However, cells with KOSR in hypoxia condition expressed significantly higher stemness markers such as C-myc and Oct4 (31.20% and 13.44% respectively) in comparison to hRPCs cultured in KOSR at normoxia (12.07% and 4.05%). Furthermore, levels of markers for retinal commitment such as rhodopsin were significantly lower in the KOSR supplemented cells in hypoxia culture compared to normoxia. KOSR is known to improve proliferation and maintain stemness of embryonic cells and our experiments suggest that hRPCs maintain their proliferation and stemness characteristics in hypoxia with KOSR supplement. Normoxia, however, results in mature cell marker expression, suggesting a profound effect of oxygen tension on these cells.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Proliferación Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero/farmacología , Oxígeno/farmacología , Retina/citología , Células Madre/efectos de los fármacos , Animales , Bovinos , Hipoxia de la Célula , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Rodopsina/metabolismo , Albúmina Sérica Bovina/farmacología , Células Madre/citología , Células Madre/metabolismo
11.
Exp Eye Res ; 185: 107688, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31185219

RESUMEN

Photoreceptor degeneration is a significant cause of visual impairment in the United States and globally. Cell replacement therapy shows great promise in restoring vision by transplanting stem-like cells into the sub-retinal space as substitutes for damaged photoreceptors. However, vision repair via transplantation has been limited, in large part, by low numbers of replacement cells able to migrate into damaged retinal tissue and integrate with native photoreceptors. Projects have used external chemical fields and applied electric fields to induce the chemotaxis and electrotaxis of replacement cells, respectively, with limited success. However, the application of combined electro-chemotactic fields in directing cells within biomaterials and host tissue has been surprisingly understudied. The current work examined the ability of combined electro-chemotactic fields to direct the migration of transplantable retinal progenitor cells (RPCs) in controlled microenvironments. Experiments used our established galvano-microfluidic system (Gal-MµS) to generate tunable chemotactic concentration fields with and without superimposed electric fields. Result illustrate that combination fields increased the distance migrated by RPCs by over three times that seen in either field, individually, and with greater directionality towards increasing gradients. Interestingly, immunofluorescence assays showed no significant differences in the distribution of the total and/or activated cognate receptor of interest, indicating that changes in ligand binding alone were not responsible for the measured increases in migration. Bioinformatics analysis was then performed to identity potential, synergistic mechanistic pathways involved in the electro-chemotaxis measured. Results indicate that increased RPC migration in electro-chemotactic fields may arise from down-regulation of cell adhesion proteins in tandem with up-regulation of cytoskeletal regulation proteins. These comprehensive results point towards a novel migration-targeted treatment that may dramatically improve transplantation outcomes as well as elucidate unreported synergy across biological mechanisms in response to electro-chemotactic fields.


Asunto(s)
Movimiento Celular/fisiología , Quimiotaxis/fisiología , Campos Electromagnéticos , Retina/citología , Trasplante de Células Madre , Células Madre/citología , Animales , Cadherinas/metabolismo , Células Cultivadas , ADN-Topoisomerasas de Tipo II/genética , Expresión Génica , Inmunohistoquímica , Dispositivos Laboratorio en un Chip , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión a Poli-ADP-Ribosa/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CXCR4/genética , Células Madre/fisiología , beta Catenina/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/genética
12.
Differentiation ; 101: 8-15, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29574166

RESUMEN

Recently, we have found that human stem cells from apical papilla (SCAP) show a stromal cell-derived inducing activity (SDIA). To examine SDIA competence for retinal cells differentiation, we co-cultured SCAP with human pluripotent stem cells (hPSCs). In comparison with Matrigel-cultured hPSCs, SCAP significantly induces hPSCs to differentiate into rostral neural cells as demonstrated by upregulation of OTX2 and PAX6 and down-regulation of EN1, HOXB4 and HOXC8. Furthermore, the differentiated cells on SCAP significantly expressed eye-field markers, RAX, PAX6, LHX2 and SIX3 and showed five folds pigmented colonies. The generated hPSC-retinal pigmented epithelium (RPE) was hexagonal and highly expressed related markers, ZO-1, RPE65, BEST, CRALBP and MITF. They were able to phagocytose latex beads. Moreover, the assessment of the isolated neural tube-like structures on SCAP showed the expression of retinal progenitor cells (RPCs) - SIX3, RAX, and PAX6. SCAP highly expressed DKK3 and SFRP2, Wnt inhibitor factors and their target genes, Cyclin D1 and c-Myc were down-regulated significantly on SCAP. These results showed SCAP promoted the differentiation of hPSCs into retinal cells (RPE and RPCs) possibly through inhibition of Wnt signaling pathway. This simple and efficient approach provides human RPE generation for developing therapies for diseases such as age-related macular degeneration.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias Humanas/citología , Células Madre Pluripotentes/citología , Retina/citología , Epitelio Pigmentado de la Retina/citología , Técnicas de Cultivo de Célula/métodos , Línea Celular , Células Cultivadas , Humanos
13.
Dev Dyn ; 247(11): 1199-1210, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30311321

RESUMEN

BACKGROUND: The retinal homeobox (rx/rax) gene is a transcription factor expressed in the developing eye field that is necessary for normal eye development. rax is necessary for retinal specification and stem cell development. The genetic program of early retinal development, including rax expression, can be induced in naïve ectoderm by activation of insulin-like growth factor (IGF) signaling. We have undertaken a microarray-based approach to identify rax-dependent IGF-induced genes. RESULTS: We identified 21 IGF-induced genes that exhibit at least a two-fold decrease in expression when rax expression is knocked down. Ten of these genes were expressed in the developing eye, eight were expressed in the ciliary marginal zone of the mature tadpole retina, and four could significantly rescue the rax knockdown phenotype. One of these, the nei endonuclease VIII-like 3 (neil3) gene, rescued the rax knockdown phenotype to a remarkable degree. We found that neil3 is necessary for normal retinal lamination and retinal neuron differentiation. CONCLUSIONS: We have identified neil3 as a component of the rax genetic pathway necessary for normal retinal progenitor cell development. neil3 is involved in the base excision DNA repair pathway, suggesting that this pathway is essential for normal rax-dependent progenitor cell development in the mature retina. Developmental Dynamics 247:1199-1210, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Proteínas del Ojo/genética , N-Glicosil Hidrolasas/genética , Proteínas de Xenopus/genética , Animales , Proteínas del Ojo/análisis , Proteínas de Homeodominio/genética , Larva/crecimiento & desarrollo , Análisis por Matrices de Proteínas , Retina/química , Retina/citología , Células Madre , Proteínas de Xenopus/análisis , Xenopus laevis/embriología
14.
J Cell Physiol ; 233(2): 1222-1235, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28480968

RESUMEN

Cell replacement is a promising therapy for degenerative diseases like age-related macular degeneration (AMD). Since the human retina lacks regeneration capacity, much attention has been directed toward persuading for cells that can differentiate into retinal neurons. In this report, we have investigated reprogramming of the human RPE cells and concerned the effect of donor age on the cellular fate as a critical determinant in reprogramming competence. We evaluated the effect of SOX2 over-expression in human neonatal and adult RPE cells in cultures. The coding region of human SOX2 gene was cloned into adeno-associated virus (AAV2) and primary culture of human neonatal/adult RPE cells were infected by recombinant virus. De-differentiation of RPE to neural/retinal progenitor cells was investigated by quantitative real-time PCR and ICC for neural/retinal progenitor cells' markers. Gene expression analysis showed 80-fold and 12-fold over-expression for SOX2 gene in infected neonatal and adult hRPE cells, respectively. The fold of increase for Nestin in neonatal and adult hRPE cells was 3.8-fold and 2.5-fold, respectively. PAX6 expression was increased threefold and 2.5-fold in neonatal/adult treated cultures. Howbeit, we could not detect rhodopsin, and CHX10 expression in neonatal hRPE cultures and expression of rhodopsin in adult hRPE cells. Results showed SOX2 induced human neonatal/adult RPE cells to de-differentiate toward retinal progenitor cells. However, the increased number of PAX6, CHX10, Thy1, and rhodopsin positive cells in adult hRPE treated cultures clearly indicated the considerable generation of neuro-retinal terminally differentiated cells.


Asunto(s)
Desdiferenciación Celular , Reprogramación Celular , Dependovirus/genética , Células Epiteliales/metabolismo , Vectores Genéticos , Epitelio Pigmentado de la Retina/metabolismo , Factores de Transcripción SOXB1/metabolismo , Células Madre/metabolismo , Transducción Genética , Transfección/métodos , Adulto , Biomarcadores/metabolismo , Células Cultivadas , Técnicas de Reprogramación Celular , Regulación del Desarrollo de la Expresión Génica , Humanos , Recién Nacido , Fenotipo , Factores de Transcripción SOXB1/genética
15.
J Cell Biochem ; 118(10): 3080-3089, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28370284

RESUMEN

Retinal pigment epithelium (RPE) cells represent a great potential to rescue degenerated cells of the damaged retina. Activation of the virtually plastic properties of RPE cells may aid in recovery of retinal degenerative disorders without the need for entire RPE sheet transplantation. Poly (2-hydroxyethyl methacrylate)(PolyHEMA) is one of the most important hydrogels in the biomaterials world. This hydrophobic polymer does not normally support attachment of mammalian cells. In the current study we investigated the effect of PolyHEMA as a cell culture substrate on the growth, differentiation, and plasticity of hRPE cells. hRPE cells were isolated from neonatal human globes and cultured on PolyHEMA and polystyrene substrates (as controls) in 24-well culture plates. DMEM/F12 was supplemented with 10% fetal bovine serum (FBS) and/or 30% human amniotic fluid (HAF) for cultured cells on polystyrene and PolyHEMA coated vessels. Morphology, rate of cell proliferation and cell death, MTT assay, immunocytochemistry and Real-Time RT-PCR were performed to investigate the effects of PolyHEMA on the growth and differentiation of cultured hRPE cells. Proliferation rate of the cells that had been cultured on PolyHEMA was reduced; PolyHEMA did not induce cell death in the hRPE cultures. hRPE cells cultured on PolyHEMA formed many giant spheroid colonies. The giant colonies were re-cultured and the presence of retinal progenitor markers and markers of hRPE cells were detected in cell cultures on PolyHEMA. PolyHEMA seems to be promising for both maintenance and de-differentiation of hRPE cells and expansion of the retinal progenitor cells from the cultures that are originated from hRPE cells. J. Cell. Biochem. 118: 3080-3089, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Desdiferenciación Celular , Células Epiteliales/metabolismo , Polihidroxietil Metacrilato/química , Epitelio Pigmentado de la Retina/metabolismo , Células Madre/metabolismo , Células Epiteliales/citología , Humanos , Epitelio Pigmentado de la Retina/citología , Células Madre/citología
16.
J Cell Sci ; 128(11): 2169-78, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25918122

RESUMEN

Definitive surface markers for retinal progenitor cells (RPCs) are still lacking. Therefore, we sorted c-Kit(+) and stage-specific embryonic antigen-4(-) (SSEA4(-)) retinal cells for further biological characterization. RPCs were isolated from human fetal retinas (gestational age of 12-14 weeks). c-Kit(+)/SSEA4(-) RPCs were sorted by fluorescence-activated cell sorting, and their proliferation and differentiation capabilities were evaluated by using immunocytochemistry and flow cytometry. The effectiveness and safety were assessed following injection of c-Kit(+)/SSEA4(-) cells into the subretina of Royal College of Surgeons (RCS) rats. c-Kit(+) cells were found in the inner part of the fetal retina. Sorted c-Kit(+)/SSEA4(-) cells expressed retinal stem cell markers. Our results clearly demonstrate the proliferative potential of these cells. Moreover, c-Kit(+)/SSEA4(-) cells differentiated into retinal cells that expressed markers of photoreceptor cells, ganglion cells and glial cells. These cells survived for at least 3 months after transplantation into the host subretinal space. Teratomas were not observed in the c-Kit(+)/SSEA4(-)-cell group. Thus, c-Kit can be used as a surface marker for RPCs, and c-Kit(+)/SSEA4(-) RPCs exhibit the ability to self-renew and differentiate into retinal cells.


Asunto(s)
Feto/citología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Retina/citología , Células Madre/citología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Feto/metabolismo , Citometría de Flujo/métodos , Humanos , Masculino , Ratones , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Células Fotorreceptoras/citología , Células Fotorreceptoras/metabolismo , Ratas , Retina/metabolismo , Degeneración Retiniana/metabolismo , Antígenos Embrionarios Específico de Estadio/metabolismo , Trasplante de Células Madre/métodos , Células Madre/metabolismo
17.
Genesis ; 54(11): 562-567, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27696680

RESUMEN

The Retinal homeobox (rax) gene is expressed in vertebrate retinal progenitor and stem cells and is essential for retinal development. In frogs, rax is expressed in the ciliary marginal zone (CMZ), a region containing retinal progenitor and stem cells at the anterior of the eye. Little is known regarding regulation of rax transcription and regulation of transcription of rax targets. We found that three ultra-conserved genomic elements (UCEs) flanking the rax coding region regulate expression of a rax promoter-GFP transgene in Xenopus tadpoles. One of these elements, UCE1, regulates expression of the transgene in the dorsal CMZ. UCE1 contains a Rax binding site, PCE-1. We demonstrate that rax regulates expression of the transgene through the PCE-1 site found in UCE1. Therefore, rax transcription in the CMZ is controlled, in part, by autoregulatory mechanisms.


Asunto(s)
Proteínas del Ojo/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Retina/crecimiento & desarrollo , Proteínas de Xenopus/genética , Xenopus/genética , Animales , Sitios de Unión , Secuencia Conservada/genética , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Larva/genética , Larva/crecimiento & desarrollo , Regiones Promotoras Genéticas , Retina/metabolismo , Xenopus/crecimiento & desarrollo , Proteínas de Xenopus/biosíntesis , Proteínas de Xenopus/metabolismo
18.
J Biol Chem ; 289(10): 6362-6371, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24407289

RESUMEN

Cell transplantation is a potential therapeutic strategy for retinal degenerative diseases involving the loss of photoreceptors. However, it faces challenges to clinical translation due to safety concerns and a limited supply of cells. Human retinal progenitor cells (hRPCs) from fetal neural retina are expandable in vitro and maintain an undifferentiated state. This study aimed to investigate the therapeutic potential of hRPCs transplanted into a Royal College of Surgeons (RCS) rat model of retinal degeneration. At 12 weeks, optokinetic response showed that hRPC-grafted eyes had significantly superior visual acuity compared with vehicle-treated eyes. Histological evaluation of outer nuclear layer (ONL) characteristics such as ONL thickness, spread distance, and cell count demonstrated a significantly greater preservation of the ONL in hRPC-treated eyes compared with both vehicle-treated and control eyes. The transplanted hRPCs arrested visual decline over time in the RCS rat and rescued retinal morphology, demonstrating their potential as a therapy for retinal diseases. We suggest that the preservation of visual acuity was likely achieved through host photoreceptor rescue. We found that hRPC transplantation into the subretinal space of RCS rats was well tolerated, with no adverse effects such as tumor formation noted at 12 weeks after treatment.


Asunto(s)
Células Madre Embrionarias/trasplante , Epitelio Pigmentado Ocular/trasplante , Retina , Degeneración Retiniana/cirugía , Trasplante de Células Madre , Animales , Separación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Feto/citología , Humanos , Ratas , Retina/citología , Retina/embriología , Retina/fisiología , Degeneración Retiniana/fisiopatología , Agudeza Visual
19.
J Theor Biol ; 373: 73-81, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25797309

RESUMEN

The vertebrate retina is made up of seven principal cell types. These seven retinal cell types arise from multipotent retinal progenitor cells (RPCs). The competency model was proposed suggesting that RPCs undergo a series of irreversible transitions between competency states, in each of which the RPCs are competent to generate a different subset of cell types, but not retinal cells generated at previous moments. In this work, we generalize the stochastic model of neurogenesis of Barton et al. (2014), assuming that the same factor that regulates the differentiation, regulates the competency. The model reproduces the timing of production of different retinal cell types in rats such as it was experimentally measured. The results show that the evolution of the competency during retinogenesis could be explained by a single factor. Its evolution during the cell cycle and the stochastic inheritance in cell divisions determine the sequence and the overlap of production of different retinal cell types during development.


Asunto(s)
Modelos Neurológicos , Retina/citología , Retina/embriología , Animales , Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Neurogénesis/fisiología , Ratas , Procesos Estocásticos
20.
Exp Eye Res ; 123: 115-20, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24811219

RESUMEN

The ciliary marginal zone (CMZ) is a circumferential ring of cells found at the extreme periphery of the maturing and mature neural retina that consists of retinal stem and progenitor cells. It functions to add retinal neurons to the periphery of the neural retina in larval and adult fish, larval frogs, and birds. Additionally, the CMZ may contribute to regeneration of the damaged retina in frogs and fish. In mammals, cells from the ciliary epithelium can be induced to express retinal stem cell-like characteristics in culture but may not comprise a classically defined CMZ.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA