Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Int J Mol Sci ; 25(3)2024 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-38338800

RESUMEN

Degenerative retinal diseases associated with photoreceptor loss are a leading cause of visual impairment worldwide, with limited treatment options. Phenotypic profiling coupled with medicinal chemistry were used to develop a small molecule with proliferative effects on retinal stem/progenitor cells, as assessed in vitro in a neurosphere assay and in vivo by measuring Msx1-positive ciliary body cell proliferation. The compound was identified as having kinase inhibitory activity and was subjected to cellular pathway analysis in non-retinal human primary cell systems. When tested in a disease-relevant murine model of adult retinal degeneration (MNU-induced retinal degeneration), we observed that four repeat intravitreal injections of the compound improved the thickness of the outer nuclear layer along with the regeneration of the visual function, as measured with ERG, visual acuity, and contrast sensitivity tests. This serves as a proof of concept for the use of a small molecule to promote endogenous regeneration in the eye.


Asunto(s)
Degeneración Retiniana , Humanos , Ratones , Animales , Degeneración Retiniana/metabolismo , Metilnitrosourea , Retina/metabolismo , Células Fotorreceptoras , Regeneración , Modelos Animales de Enfermedad , Mamíferos
2.
Dev Biol ; 445(2): 256-270, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30472119

RESUMEN

The enteric nervous system is thought to originate solely from the neural crest. Transgenic lineage tracing revealed a novel population of clonal pancreatic duodenal homeobox-1 (Pdx1)-Cre lineage progenitor cells in the tunica muscularis of the gut that produced pancreatic descendants as well as neurons upon differentiation in vitro. Additionally, an in vivo subpopulation of endoderm lineage enteric neurons, but not glial cells, was seen especially in the proximal gut. Analysis of early transgenic embryos revealed Pdx1-Cre progeny (as well as Sox-17-Cre and Foxa2-Cre progeny) migrating from the developing pancreas and duodenum at E11.5 and contributing to the enteric nervous system. These results show that the mammalian enteric nervous system arises from both the neural crest and the endoderm. Moreover, in adult mice there are separate Wnt1-Cre neural crest stem cells and Pdx1-Cre pancreatic progenitors within the muscle layer of the gut.


Asunto(s)
Sistema Nervioso Entérico/embriología , Animales , Linaje de la Célula/genética , Duodeno/embriología , Duodeno/inervación , Duodeno/metabolismo , Endodermo/citología , Endodermo/embriología , Endodermo/metabolismo , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Páncreas/embriología , Páncreas/inervación , Páncreas/metabolismo , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
3.
Stem Cells ; 31(10): 2218-30, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23843349

RESUMEN

Rare retinal stem cells (RSCs) within the ciliary epithelium at the retinal margin of the adult mouse and human eyes can divide in vitro in the absence of growth factors to generate clonal, self-renewing spheres which can generate all the retinal cell types. Since no regenerative properties are seen in situ in the adult mammalian eye, we sought to determine the factors that are involved in the repression of endogenous RSCs. We discovered that factors secreted by the adult lens and cornea block the proliferation of adult RSCs in vitro. Bone morphogenetic protein (BMP)2, BMP4, and secreted frizzled related protein 2 were identified as principal effectors of the anti-proliferative effects on RSCs. As a similar induced quiescence was observed in vitro on both mouse and human RSCs, targeting these molecules in vivo may reactivate RSCs directly in situ in the eyes of the blind.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Proliferación Celular , Proteínas de la Membrana/fisiología , Células Madre/fisiología , Animales , Proteína Morfogenética Ósea 2/fisiología , Proteína Morfogenética Ósea 4/fisiología , Proteínas Portadoras , Células Cultivadas , Técnicas de Cocultivo , Córnea/metabolismo , Cristalino/metabolismo , Ratones , Ratones Endogámicos C57BL , Retina/citología , Esferoides Celulares/fisiología , Técnicas de Cultivo de Tejidos
4.
Stem Cells Dev ; 32(19-20): 606-621, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37551982

RESUMEN

The mature brain contains an incredible number and diversity of cells that are produced and maintained by heterogeneous pools of neural stem cells (NSCs). Two distinct types of NSCs exist in the developing and adult mouse brain: Glial Fibrillary Acidic Protein (GFAP)-negative primitive (p)NSCs and downstream GFAP-positive definitive (d)NSCs. To better understand the embryonic functions of NSCs, we performed clonal lineage tracing within neurospheres grown from either pNSCs or dNSCs to enrich for their most immediate downstream neural progenitor cells (NPCs). These clonal progenitor lineage tracing data allowed us to construct a hierarchy of progenitor subtypes downstream of pNSCs and dNSCs that were then validated using single-cell transcriptomics. Further, we identify Nexn as required for neuronal specification from neuron/astrocyte progenitor cells downstream of rare pNSCs. Combined, these data provide single-cell resolution of NPC lineages downstream of rare pNSCs that likely would be missed from population-level analyses in vivo.


Asunto(s)
Células-Madre Neurales , Ratones , Animales , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Encéfalo/metabolismo , Astrocitos/metabolismo , Diferenciación Celular/genética
5.
Stem Cells ; 28(3): 489-500, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20014120

RESUMEN

Retinal stem cells (RSCs) are present in the ciliary margin of the adult human eye and can give rise to all retinal cell types. Here we show that modulation of retinal transcription factor gene expression in human RSCs greatly enriches photoreceptor progeny, and that strong enrichment was obtained with the combined transduction of OTX2 and CRX together with the modulation of CHX10. When these genetically modified human RSC progeny are transplanted into mouse eyes, their retinal integration and differentiation is superior to unmodified RSC progeny. Moreover, electrophysiologic and behavioral tests show that these transplanted cells promote functional recovery in transducin mutant mice. This study suggests that gene modulation in human RSCs may provide a source of photoreceptor cells for the treatment of photoreceptor disease.


Asunto(s)
Diferenciación Celular/genética , Células Fotorreceptoras de Vertebrados/citología , Retina/citología , Trasplante de Células Madre/métodos , Células Madre/citología , Trasplante Heterólogo/métodos , Animales , Linaje de la Célula/genética , Células Cultivadas , Regulación de la Expresión Génica/genética , Supervivencia de Injerto/genética , Proteínas de Homeodominio/genética , Humanos , Ratones , Factores de Transcripción Otx/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/metabolismo , Células Madre/metabolismo , Transactivadores/genética , Factores de Transcripción/genética , Transducina/genética , Transducción Genética/métodos , Transfección/métodos
6.
Stem Cell Res Ther ; 12(1): 560, 2021 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-34717744

RESUMEN

BACKGROUND: The adult mammalian retina does not have the capacity to regenerate cells lost due to damage or disease. Therefore, retinal injuries and blinding diseases result in irreversible vision loss. However, retinal stem cells (RSCs), which participate in retinogenesis during development, persist in a quiescent state in the ciliary epithelium (CE) of the adult mammalian eye. Moreover, RSCs retain the ability to generate all retinal cell types when cultured in vitro, including photoreceptors. Therefore, it may be possible to activate endogenous RSCs to induce retinal neurogenesis in vivo and restore vision in the adult mammalian eye. METHODS: To investigate if endogenous RSCs can be activated, we performed combinatorial intravitreal injections of antagonists to BMP and sFRP2 proteins (two proposed mediators of RSC quiescence in vivo), with or without growth factors FGF and Insulin. We also investigated the effects of chemically-induced N-methyl-N-Nitrosourea (MNU) retinal degeneration on RSC activation, both alone and in combination withthe injected factors. Further, we employed inducible Msx1-CreERT2 genetic lineage labeling of the CE followed by stimulation paradigms to determine if activated endogenous RSCs could migrate into the retina and differentiate into retinal neurons. RESULTS: We found that in vivo antagonism of BMP and sFRP2 proteins induced CE cells in the RSC niche to proliferate and expanded the RSC population. BMP and sFRP2 antagonism also enhanced CE cell proliferation in response to exogenous growth factor stimulation and MNU-induced retinal degeneration. Furthermore, Msx1-CreERT2 genetic lineage tracing revealed that CE cells migrated into the retina following stimulation and/or injury, where they expressed markers of mature photoreceptors and retinal ganglion cells. CONCLUSIONS: Together, these results indicate that endogenous adult mammalian RSCs may have latent regenerative potential that can be activated by modulating the RSC niche and hold promise as a means for endogenous retinal cell therapy to repair the retina and improve vision.


Asunto(s)
Retina , Células Madre , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Mamíferos , Retina/metabolismo , Células Madre/metabolismo
7.
Stem Cell Res Ther ; 12(1): 83, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33494791

RESUMEN

BACKGROUND: Adult mammalian retinal stem cells (RSCs) readily proliferate, self-renew, and generate progeny that differentiate into all retinal cell types in vitro. RSC-derived progeny can be induced to differentiate into photoreceptors, making them a potential source for retinal cell transplant therapies. Despite their proliferative propensity in vitro, RSCs in the adult mammalian eye do not proliferate and do not have a regenerative response to injury. Thus, identifying and modulating the mechanisms that regulate RSC proliferation may enhance the capacity to produce RSC-derived progeny in vitro and enable RSC activation in vivo. METHODS: Here, we used medium-throughput screening to identify small molecules that can expand the number of RSCs and their progeny in culture. In vitro differentiation assays were used to assess the effects of synthetic glucocorticoid agonist dexamethasone on RSC-derived progenitor cell fate. Intravitreal injections of dexamethasone into adult mouse eyes were used to investigate the effects on endogenous RSCs. RESULTS: We discovered that high-affinity synthetic glucocorticoid agonists increase RSC self-renewal and increase retinal progenitor proliferation up to 6-fold without influencing their differentiation in vitro. Intravitreal injection of synthetic glucocorticoid agonist dexamethasone induced in vivo proliferation in the ciliary epithelium-the niche in which adult RSCs reside. CONCLUSIONS: Together, our results identify glucocorticoids as novel regulators of retinal stem and progenitor cell proliferation in culture and provide evidence that GCs may activate endogenous RSCs.


Asunto(s)
Autorrenovación de las Células , Glucocorticoides , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Glucocorticoides/farmacología , Ratones , Retina
8.
Lab Chip ; 21(22): 4464-4476, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34651637

RESUMEN

Loss of photoreceptors due to retinal degeneration is a major cause of untreatable visual impairment and blindness. Cell replacement therapy, using retinal stem cell (RSC)-derived photoreceptors, holds promise for reconstituting damaged cell populations in the retina. One major obstacle preventing translation to the clinic is the lack of validated markers or strategies to prospectively identify these rare cells in the retina and subsequently enrich them. Here, we introduce a microfluidic platform that combines nickel micromagnets, herringbone structures, and a design enabling varying flow velocities among three compartments to facilitate a highly efficient enrichment of RSCs. In addition, we developed an affinity enrichment strategy based on cell-surface markers that was utilized to isolate RSCs from the adult ciliary epithelium. We showed that targeting a panel of three cell surface markers simultaneously facilitates the enrichment of RSCs to 1 : 3 relative to unsorted cells. Combining the microfluidic platform with single-cell whole-transcriptome profiling, we successfully identified four differentially expressed cell surface markers that can be targeted simultaneously to yield an unprecedented 1 : 2 enrichment of RSCs relative to unsorted cells. We also identified transcription factors (TFs) that play functional roles in maintenance, quiescence, and proliferation of RSCs. This level of analysis for the first time identified a spectrum of molecular and functional properties of RSCs.


Asunto(s)
Microfluídica , Retina , Animales , Diferenciación Celular , Proliferación Celular , Perfilación de la Expresión Génica , Ratones , Células Madre
9.
ACS Appl Mater Interfaces ; 10(41): 34811-34816, 2018 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-30265796

RESUMEN

Retinal stem cells (RSCs) are promising candidates for patient-derived cell therapy to repair damage to the eye; however, RSCs are rare in retinal samples and lack validated markers, making cell sorting a significant challenge. Here we report a high-resolution deterministic lateral displacement microfluidic device that profiles RSCs in distinct size populations. Only by developing a chip that promotes cell tumbling do we limit cell deformation through apertured channels and thereby increase the size-sorting resolution of the device. We systematically explore a spectrum of microstructures, including optimized notched pillars, to study and then rationally promote cell tumbling. We find that RSCs exhibit larger diameters than most ciliary epithelial cells, an insight into RSC morphology that allows enrichment from biological samples.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Epiteliales/metabolismo , Dispositivos Laboratorio en un Chip , Retina/metabolismo , Células Madre/metabolismo , Animales , Células Cultivadas , Células Epiteliales/citología , Humanos , Ratones , Retina/citología , Células Madre/citología
10.
Stem Cell Res ; 33: 215-227, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30453152

RESUMEN

During development, multipotent progenitors undergo temporally-restricted differentiation into post-mitotic retinal cells; however, the mechanisms of progenitor division that occurs during retinogenesis remain controversial. Using clonal analyses (lineage tracing and single cell cultures), we identify rod versus cone lineage-specific progenitors derived from both adult retinal stem cells and embryonic neural retinal precursors. Taurine and retinoic acid are shown to act in an instructive and lineage-restricted manner early in the progenitor lineage hierarchy to produce rod-restricted progenitors from stem cell progeny. We also identify an instructive, but lineage-independent, mechanism for the specification of cone-restricted progenitors through the suppression of multiple differentiation signaling pathways. These data indicate that exogenous signals play critical roles in directing lineage decisions and resulting in fate-restricted rod or cone photoreceptor progenitors in culture. Additional factors may be involved in governing photoreceptor fates in vivo.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Retina/fisiopatología , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular , Ratones
11.
Stem Cell Res ; 21: 141-147, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28494434

RESUMEN

Adult retinal stem cells (RSCs) are rare quiescent cells within the ciliary epithelium of the eye, which is made up of non-pigmented N-Cadherin+ve inner and pigmented P-Cadherin+ve outer cell layers. Through FACs and single cell analyses, we have shown that RSCs arise from single cells from within the pigmented CE and express P-Cadherin. However, whether the expression of P-Cadherin is required for maintenance of the stem cell in vivo or in the formation of the clonal stem cell spheres in vitro is not known. Using cadherin functional blocking antibody experiments and a P-Cadherin -/- mouse to test whether the RSC population is affected by the loss of P-Cadherin expression, our experiments demonstrate that the RSCs reside in the pigmented CE layer and express P-Cadherin, which is important to the formation of adherent sphere colonies in vitro, however P-Cadherin is not required for maintenance of RSCs in vivo.


Asunto(s)
Cadherinas/metabolismo , Retina/citología , Células Madre/citología , Células Madre/metabolismo , Animales , Anticuerpos Bloqueadores/farmacología , Cilios/metabolismo , Células Clonales , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Ratones Endogámicos C57BL , Pigmentación , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Nicho de Células Madre/efectos de los fármacos , Células Madre/efectos de los fármacos
12.
Stem Cell Reports ; 4(6): 1031-45, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-25981414

RESUMEN

The utility of stem cells and their progeny in adult transplantation models has been limited by poor survival and integration. We designed an injectable and bioresorbable hydrogel blend of hyaluronan and methylcellulose (HAMC) and tested it with two cell types in two animal models, thereby gaining an understanding of its general applicability for enhanced cell distribution, survival, integration, and functional repair relative to conventional cell delivery in saline. HAMC improves cell survival and integration of retinal stem cell (RSC)-derived rods in the retina. The pro-survival mechanism of HAMC is ascribed to the interaction of the CD44 receptor with HA. Transient disruption of the retinal outer limiting membrane, combined with HAMC delivery, results in significantly improved rod survival and visual function. HAMC also improves the distribution, viability, and functional repair of neural stem and progenitor cells (NSCs). The HAMC delivery system improves cell transplantation efficacy in two CNS models, suggesting broad applicability.


Asunto(s)
Ácido Hialurónico/química , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Trasplante de Células Madre , Células Madre/citología , Animales , Ceguera/genética , Ceguera/terapia , Ceguera/veterinaria , Supervivencia Celular , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Inmunohistoquímica , Metilcelulosa/química , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/citología , Retina/metabolismo , Células Fotorreceptoras Retinianas Bastones/citología , Rodopsina/metabolismo , Células Madre/metabolismo , Accidente Cerebrovascular/inducido químicamente , Accidente Cerebrovascular/terapia , Accidente Cerebrovascular/veterinaria
13.
Biol Open ; 1(3): 237-46, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23213414

RESUMEN

Self-renewing, multipotential retinal stem cells (RSCs) reside in the pigmented ciliary epithelium of the peripheral retina in adult mammals. RSCs can give rise to rhodopsin positive-cells, which can integrate into early postnatal retina, and represent a potentially useful option for cellular therapy. The ability to purify a stem cell population and direct the differentiation toward a particular cell lineage is a challenge facing the application of stem cells in regenerative medicine. Here we use cell sorting to prospectively enrich mouse RSCs based on size, granularity and low expression of P-cadherin and demonstrate that only rare cells with defined properties proliferate to form colonies. We show that clonally-derived mouse and human RSC progeny are multipotent and can differentiate into mature rhodopsin-positive cells with high efficiency using combinations of exogenous culture additives known to influence neural retinal development, including taurine and retinoic acid. This directed RSC differentiation follows the temporal sequence of photoreceptor differentiation in vivo, and the cells exhibit morphology, protein and gene expression consistent with primary cultures of rods in vitro. These results demonstrate that the RSC, an adult stem cell, can be enriched and directed to produce photoreceptors as a first step toward a targeted cell replacement strategy to treat retinal degenerative disease.

14.
J Vis Exp ; (43)2010 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22157975

RESUMEN

The adult mouse retinal stem cell (RSC) is a rare quiescent cell found within the ciliary epithelium (CE) of the mammalian eye(1,2,3). The CE is made up of non-pigmented inner and pigmented outer cell layers, and the clonal RSC colonies that arise from a single pigmented cell from the CE are made up of both pigmented and non-pigmented cells which can be differentiated to form all the cell types of the neural retina and the RPE. There is some controversy about whether all the cells within the spheres all contain at least some pigment(4); however the cells are still capable of forming the different cell types found within the neural retina(1-3). In some species, such as amphibians and fish, their eyes are capable of regeneration after injury(5), however; the mammalian eye shows no such regenerative properties. We seek to identify the stem cell in vivo and to understand the mechanisms that keep the mammalian retinal stem cells quiescent(6-8), even after injury as well as using them as a potential source of cells to help repair physical or genetic models of eye injury through transplantation(9-12). Here we describe how to isolate the ciliary epithelial cells from the mouse eye and grow them in culture in order to form the clonal retinal stem cell spheres. Since there are no known markers of the stem cell in vivo, these spheres are the only known way to prospectively identify the stem cell population within the ciliary epithelium of the eye.


Asunto(s)
Cuerpo Ciliar/citología , Técnicas Citológicas/métodos , Retina/citología , Animales , Células Epiteliales/citología , Ratones
15.
Dev Biol ; 308(1): 54-67, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17574231

RESUMEN

The epithelial layers of the ciliary body (CB) and iris are non-neural structures that differentiate from the anterior region of the eyecup, the ciliary margin (CM). We show here that activation of the canonical Wnt signaling pathway is sufficient and necessary for the normal development of anterior eye structures. Pharmacological activation of beta-catenin signaling with lithium (Li(+)) treatment in retinal explants in vitro induced the ectopic expression of the CM markers Otx1 and Msx1. Cre-mediated stabilization of beta-catenin expression in the peripheral retina in vivo induced a cell autonomous upregulation of CM markers at the expense of neural retina (NR) markers and inhibited neurogenesis. Consistent with a cell autonomous conversion to peripheral eye fates, the proliferation index in the region of the retina that expressed stabilized beta-catenin was identical to the wild-type CM and there was an expansion of CB-like structures at later stages. Conversely, Cre-mediated inactivation of beta-catenin reduced CM marker expression as well as the size of the CM and CB/iris. Aberrant CB development in both mouse models was also associated with a reduction in the number of retinal stem cells in vitro. In summary, activation of canonical Wnt signaling is sufficient to promote the development of peripheral eyecup fates at the expense of the NR and is also required for the normal development of anterior eyecup structures.


Asunto(s)
Retina/embriología , Proteínas Wnt/metabolismo , Animales , Secuencia de Bases , Cuerpo Ciliar/embriología , Cuerpo Ciliar/metabolismo , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Técnicas In Vitro , Operón Lac , Ratones , Ratones Transgénicos , Microftalmía/embriología , Microftalmía/genética , Microftalmía/metabolismo , Retina/metabolismo , Transducción de Señal , Proteínas Wnt/genética , beta Catenina/deficiencia , beta Catenina/genética , beta Catenina/metabolismo
16.
Dev Biol ; 304(2): 713-21, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17316600

RESUMEN

Retinal stem cells (RSCs) exist as rare pigmented ciliary epithelial cells in adult mammalian eyes. We hypothesized that RSCs are at the top of the retinal cell lineage. Thus, genes expressed early in embryonic development to establish the retinal field in forebrain neuroectoderm may play important roles in RSCs. Pax6, a paired domain and homeodomain-containing transcription factor, is one of the earliest genes expressed in the eye field and is considered a master control gene for retinal and eye development. Here, we demonstrate that Pax6 is enriched in RSCs. Inactivation of Pax6 in vivo results in loss of competent RSCs as assayed by the failure to form clonal RSC spheres from the optic vesicles of conventional Pax6 knockout embryos and from the ciliary epithelial cells of adult Pax6 conditional knockout mice. In vitro clonal inactivation of Pax6 in adult RSCs results in a serious proliferation defect, suggesting that Pax6 is required for the proliferation and expansion of RSCs.


Asunto(s)
Proliferación Celular , Células Epiteliales/citología , Proteínas del Ojo/fisiología , Proteínas de Homeodominio/fisiología , Factores de Transcripción Paired Box/fisiología , Proteínas Represoras/fisiología , Retina/citología , Células Madre/citología , Animales , Células Epiteliales/metabolismo , Proteínas del Ojo/genética , Proteínas de Homeodominio/genética , Ratones , Ratones Noqueados , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Proteínas Represoras/genética , Retina/embriología , Retina/metabolismo , Células Madre/metabolismo
17.
Eur J Neurosci ; 23(1): 75-82, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16420417

RESUMEN

Retinal stem cells [with the potential to produce either neural retinal progenitors or retinal pigment epithelial (RPE) progenitors] exist in the mammalian eye throughout life, and indeed the greatest absolute increase in the stem population occurs postnatally. The stem cells proliferate embryonically and thus may help to build the retina initially, but in postnatal mammals they clearly do not proliferate to regenerate the retina in response to injury. Using Chx10(orJ/orJ) and Mitf(mi/mi) mice, with small eye phenotypes due to the reduction of the neural retinal progenitor population and the retinal pigmented epithelial progenitor population, respectively, we now report that the retinal stem cell population, when assayed from the ciliary margin, increases 3-8-fold in both mutants. These findings suggest that the mammalian retinal stem cell population may be capable of responding to genetically induced signals from the progenitor populations.


Asunto(s)
Ojo , Epitelio Pigmentado Ocular/fisiología , Retina/citología , Células Madre/fisiología , Animales , Animales Recién Nacidos , Recuento de Células/métodos , Diferenciación Celular/genética , Embrión de Mamíferos , Ojo/citología , Ojo/embriología , Ojo/crecimiento & desarrollo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Mutantes , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Modelos Biológicos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Proc Natl Acad Sci U S A ; 101(44): 15772-7, 2004 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-15505221

RESUMEN

This study identifies and characterizes retinal stem cells (RSCs) in early postnatal to seventh-decade human eyes. Different subregions of human eyes were dissociated and cultured by using a clonal sphere-forming assay. The stem cells were derived only from the pars plicata and pars plana of the retinal ciliary margin, at a frequency of approximately 1:500. To test for long-term self-renewal, both the sphere assay and monolayer passaging were used. By using the single sphere passaging assay, primary spheres were dissociated and replated, and individual spheres demonstrated 100% self-renewal, with single spheres giving rise to one or more new spheres in each subsequent passage. The clonal retinal spheres were plated under differentiation conditions to assay the differentiation potential of their progeny. The spheres were produced all of the different retinal cell types, demonstrating multipotentiality. Therefore, the human eye contains a small population of cells (approximately equal to 10,000 cells per eye) that have retinal stem-cell characteristics (proliferation, self-renewal, and multipotentiality). To test the in vivo potential of the stem cells and their progeny, we transplanted dissociated human retinal sphere cells, containing both stem cells and progenitors, into the eyes of postnatal day 1 NOD/SCID mice and embryonic chick eyes. The progeny of the RSCs were able to survive, migrate, integrate, and differentiate into the neural retina, especially as photoreceptors. Their facile isolation, integration, and differentiation suggest that human RSCs eventually may be valuable in treating human retinal diseases.


Asunto(s)
Células Madre Multipotentes/citología , Retina/química , Adolescente , Adulto , Anciano , Animales , Diferenciación Celular , Separación Celular/métodos , Embrión de Pollo , Niño , Preescolar , Humanos , Técnicas In Vitro , Lactante , Recién Nacido , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Células Madre Multipotentes/trasplante , Trasplante de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA