Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Invest Ophthalmol Vis Sci ; 58(1): 299-308, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28114591

RESUMEN

Purpose: The role of ß-adrenergic receptor (AR) signaling in neovascular ocular diseases has recently emerged. We have previously reported that intraperitoneal propranolol inhibits choroidal neovascularization (CNV) in vivo and ß2-AR blockade reduces vascular endothelial growth factor (VEGF) expression in mouse retinal pigment epithelium and choroidal endothelial cells in culture. Here we tested the hypothesis that the ß2-AR regulates CNV through modulation of VEGF and inflammatory cytokine expression. Methods: Mice were subjected to laser burns, inducing CNV, and were treated with an intravitreal ß2-AR antagonist. After 3 and 5 days, total eye interleukin-6 (IL-6) and VEGF protein levels were measured, respectively. After 14 days, CNV was measured on choroidal-scleral flatmounts. The effects of ß-AR signaling on VEGF and IL-6 expression were investigated in various mouse retinal and human RPE cells by using specific ß-AR agonists and antagonists. Results: ß2-Adrenergic receptor signaling increased Vegf mRNA expression by approximately 3- to 4-fold in mouse retinal microglia and pericytes in culture. ß2-Adrenergic receptor signaling upregulated IL-6 mRNA expression between 10- and 60-fold in mouse retinal microglia, pericytes, RPE, and choroidal endothelial cells in culture. Intravitreal injection of ß2-AR antagonist ICI 118,551 reduced CNV by 35% and decreased IL-6 protein levels by approximately 50%. In primary human RPE cells, ß2-AR activation also stimulated VEGF and IL-6 mRNA expression by 2- and 10-fold, respectively. Conclusions: Anti-VEGF therapy for CNV is highly effective; however, some patients are resistant to therapy while others undergo repeated, frequent treatments. ß2-Adrenergic receptor signaling is a potential therapeutic target because of its angiogenic and inflammatory properties.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Neovascularización Coroidal/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-6/genética , ARN Mensajero/genética , Epitelio Pigmentado de la Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Células Cultivadas , Coroides/efectos de los fármacos , Coroides/metabolismo , Coroides/patología , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/biosíntesis , Ratones , Ratones Endogámicos C57BL , Receptores Adrenérgicos beta/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/patología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/biosíntesis
2.
Stem Cells ; 34(11): 2625-2634, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27301076

RESUMEN

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


Asunto(s)
Tipificación del Cuerpo/genética , Proteínas de Homeodominio/genética , Células Madre Pluripotentes Inducidas/metabolismo , Microftalmía/genética , Epitelio Pigmentado de la Retina/metabolismo , Factores de Transcripción/genética , Proteína Wnt1/genética , Sustitución de Aminoácidos , Benzotiazoles/farmacología , Biomarcadores/metabolismo , Diferenciación Celular , Cuerpos Embrioides/efectos de los fármacos , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/patología , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Microftalmía/metabolismo , Microftalmía/patología , Mutación , Fenotipo , Cultivo Primario de Células , Piridinas/farmacología , Pirimidinas/farmacología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/patología , Factores de Transcripción/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt1/agonistas , Proteína Wnt1/antagonistas & inhibidores , Proteína Wnt1/metabolismo
3.
PLoS One ; 10(8): e0135830, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26292211

RESUMEN

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


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Proteínas de Homeodominio/fisiología , Células-Madre Neurales/fisiología , Retina/citología , Factores de Transcripción/fisiología , Diferenciación Celular/fisiología , Humanos , Imagenología Tridimensional , Neurogénesis/fisiología , Reacción en Cadena de la Polimerasa , Retina/embriología , Retina/fisiología
4.
Hum Mol Genet ; 23(23): 6332-44, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25008112

RESUMEN

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


Asunto(s)
Células Madre Embrionarias/metabolismo , Factor de Transcripción Asociado a Microftalmía/genética , Células-Madre Neurales/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Células Madre Embrionarias/citología , Técnicas de Inactivación de Genes , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Factor de Transcripción Asociado a Microftalmía/metabolismo , Células-Madre Neurales/citología , Regiones Promotoras Genéticas , Isoformas de Proteínas/metabolismo , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/embriología , Factores de Transcripción/metabolismo
5.
Exp Eye Res ; 123: 161-72, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24534198

RESUMEN

Human pluripotent stem cells have made a remarkable impact on science, technology and medicine by providing a potentially unlimited source of human cells for basic research and clinical applications. In recent years, knowledge gained from the study of human embryonic stem cells and mammalian somatic cell reprogramming has led to the routine production of human induced pluripotent stem cells (hiPSCs) in laboratories worldwide. hiPSCs show promise for use in transplantation, high throughput drug screening, "disease-in-a-dish" modeling, disease gene discovery, and gene therapy testing. This review will focus on the first application, beginning with a discussion of methods for producing retinal lineage cells that are lost in inherited and acquired forms of retinal degenerative disease. The selection of appropriate hiPSC-derived donor cell type(s) for transplantation will be discussed, as will the caveats and prerequisite steps to formulating a clinical Good Manufacturing Practice (cGMP) product for clinical trials.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Degeneración Retiniana/terapia , Trasplante de Células Madre , Linaje de la Célula , Humanos
6.
Stem Cells ; 32(6): 1480-92, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24532057

RESUMEN

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


Asunto(s)
Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Retina/embriología , Retina/metabolismo , Factores de Transcripción/metabolismo , Adulto , Sustitución de Aminoácidos , Animales , Tipificación del Cuerpo/genética , Diferenciación Celular , Línea Celular , Linaje de la Célula , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Masculino , Ratones , Mutación/genética , Fenotipo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Retina/patología , Células Bipolares de la Retina/metabolismo , Células Bipolares de la Retina/patología , Epitelio Pigmentado de la Retina/embriología , Epitelio Pigmentado de la Retina/patología , Análisis de Secuencia de ARN , Transducción de Señal/genética , Factores de Transcripción/genética , Transcriptoma/genética
7.
Invest Ophthalmol Vis Sci ; 54(10): 6767-78, 2013 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-24030465

RESUMEN

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


Asunto(s)
Células Madre Embrionarias/citología , Epitelio Pigmentado de la Retina/embriología , Animales , Western Blotting , Bovinos , Diferenciación Celular , Línea Celular , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunohistoquímica , Fagocitosis , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Epitelio Pigmentado de la Retina/metabolismo
9.
Hum Mol Genet ; 22(3): 593-607, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23139242

RESUMEN

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


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/fisiopatología , Animales , Bestrofinas , Calcio/metabolismo , Bovinos , Diferenciación Celular , Línea Celular , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Regulación de la Expresión Génica , Homeostasis , Humanos , Inmunohistoquímica , Inmunoprecipitación , Mácula Lútea/patología , Microscopía Electrónica de Transmisión , Estrés Oxidativo , Fagocitosis , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/patología , Distrofia Macular Viteliforme/metabolismo
10.
Invest Ophthalmol Vis Sci ; 53(4): 2007-19, 2012 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-22410558

RESUMEN

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


Asunto(s)
Células Madre Pluripotentes Inducidas/fisiología , Morfogénesis , Retina/crecimiento & desarrollo , Sinapsis/fisiología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/citología , Retina/citología , Retina/metabolismo
11.
Stem Cells ; 29(8): 1206-18, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21678528

RESUMEN

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


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Células Madre Pluripotentes/fisiología , Enfermedades de la Retina/terapia , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Expresión Génica , Terapia Genética , Atrofia Girata/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Potenciales de la Membrana , Técnicas de Placa-Clamp , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Células Fotorreceptoras/fisiología , Medicina de Precisión , Prosencéfalo/embriología , Retina/embriología , Retina/patología , Epitelio Pigmentado de la Retina/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Proc Natl Acad Sci U S A ; 106(39): 16698-703, 2009 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-19706890

RESUMEN

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


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Pluripotentes/citología , Retina/crecimiento & desarrollo , Diferenciación Celular , Células Madre Embrionarias/metabolismo , Humanos , Inmunohistoquímica , Modelos Biológicos , Fenotipo , Células Madre Pluripotentes/metabolismo , Retina/embriología
13.
Stem Cells ; 26(12): 3182-93, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18802035

RESUMEN

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


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Neuronas/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Apoptosis , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Medios de Cultivo Condicionados/metabolismo , Humanos , Fenotipo , Fosforilación , Retina/citología , Células Madre/citología , Factores de Transcripción/metabolismo
14.
Invest Ophthalmol Vis Sci ; 49(7): 3201-6, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18579765

RESUMEN

PURPOSE: As a follow-up to previous studies showing that human cortical neural progenitor cells (hNPC(ctx)) can sustain vision for at least 70 days after injection into the subretinal space of Royal College of Surgeons (RCS) rats, the authors examined how functional rescue is preserved over long periods and how this relates to retinal integrity and donor cell survival. METHODS: Pigmented dystrophic RCS rats (n = 15) received unilateral subretinal injections of hNPC(ctx) at postnatal day (P) 21; control rats (n = 10) received medium alone and were untreated. All animals were maintained on oral cyclosporine A. Function was monitored serially by measuring acuity (using an optomotor test) and luminance thresholds (recording from the superior colliculus) at approximately P90, P150, and P280. Eyes were processed for histologic study after functional tests. RESULTS: Acuity and luminance thresholds were significantly better in hNPC(ctx)-treated animals than in controls (P < 0.001) at all time points studied. Acuity was greater than 90%, 82%, and 37% of normal at P90, P150, and P270, whereas luminance thresholds in the area of best rescue remained similar the whole time. Histologic studies revealed substantial photoreceptor rescue, even up to P280, despite progressive deterioration in rod and cone morphology. Donor cells were still present at P280, and no sign of donor cell overgrowth was seen. CONCLUSIONS: Long-term rescue of function and associated morphologic substrates was seen, together with donor cell survival even in the xenograft paradigm. This is encouraging when exploring further the potential for the application of hNPC(ctx) in treating retinal disease.


Asunto(s)
Neuronas/trasplante , Células Fotorreceptoras de Vertebrados , Degeneración Retiniana/fisiopatología , Degeneración Retiniana/cirugía , Trasplante de Células Madre , Trasplante Heterólogo , Visión Ocular , Animales , Supervivencia Celular , Humanos , Inyecciones , Luz , Células Fotorreceptoras de Vertebrados/patología , Ratas , Células Fotorreceptoras Retinianas Conos/patología , Degeneración Retiniana/patología , Células Fotorreceptoras Retinianas Bastones/patología , Umbral Sensorial , Trasplante de Células Madre/métodos , Factores de Tiempo , Agudeza Visual
15.
Invest Ophthalmol Vis Sci ; 49(2): 788-99, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18235029

RESUMEN

PURPOSE: Established techniques for culturing primary human retinal pigment epithelial (RPE) cells have facilitated the laboratory investigation of this multipurpose retinal cell layer. However, most culture methods involve the use of animal serum to establish and maintain RPE monolayers, which can complicate efforts to define and study factors involved in the maturation and function of these cells. Therefore, this study was conducted to develop a simple, serum-free system to propagate and sustain human RPE in vitro. METHODS: RPE was dissected from human prenatal donor eyes and cultured in serum-free defined medium containing the commercially formulated supplement B27 or N2. Cultures were grown initially as adherent tissue sections or suspended spherical aggregates and later expanded and maintained as monolayers. PCR, Western blot analysis, and immunocytochemistry were used to monitor gene and protein expression in established cultures, followed by examination of secretory products in RPE conditioned medium by ELISA and mass spectrometric analysis. RESULTS: In medium supplemented with B27, but not N2, RPE could be expanded up to 40,000-fold over six passages and maintained in culture for more than 1 year. In long-term cultures, typical cellular morphology and pigmentation were observed, along with expression of characteristic RPE markers. RPE monolayers also retained proper apical-basal orientation and secreted multiple factors implicated in the maintenance of photoreceptor health and the pathogenesis of age-related macular degeneration. CONCLUSIONS: Monolayer cultures of human prenatal RPE can be grown and maintained long term in the total absence of serum and still retain the phenotype, gene and protein expression profile, and secretory capacity exhibited by mature RPE cells.


Asunto(s)
Técnicas de Cultivo de Célula , Feto/citología , Epitelio Pigmentado Ocular/citología , Biomarcadores/metabolismo , Western Blotting , Polaridad Celular , Proliferación Celular , Medio de Cultivo Libre de Suero , Electroforesis en Gel Bidimensional , Ensayo de Inmunoadsorción Enzimática , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/genética , Técnica del Anticuerpo Fluorescente Indirecta , Expresión Génica , Edad Gestacional , Humanos , Espectrometría de Masas , Epitelio Pigmentado Ocular/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Uniones Estrechas , Donantes de Tejidos
16.
J Toxicol Environ Health A ; 70(14): 1203-13, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17573634

RESUMEN

Four groups of Fischer Brown Norway hybrid rats were exposed for 5, 10, 15, or 20 d to aerosolized-vapor jet propulsion fuel 8 (JP-8) compared to freely moving (5 and 10-d exposures) or sham-confined controls (15 and 20-d exposures). Behavioral testing utilized the U.S. Environmental Protection Agency Functional Observational Battery. Exploratory ethological factor analysis identified three salient factors (central nervous system [CNS] excitability, autonomic 1, and autonomic 2) for use in profiling JP-8 exposure in future studies. The factors were used as dependent variables in general linear modeling. Exposed animals were found to engage in more rearing and hyperaroused behavior compared to controls, replicating prior JP-8 exposure findings. Exposed animals also showed increasing but rapidly decelerating stool output (autonomic 1), and a significant increasing linear trend for urine output (autonomic 2). No significant trends were noted for either of the control groups for the autonomic factors. Rats from each of the groups for each of the time frames were randomly selected for tissue assay from seven brain regions for neurotransmitter levels. Hippocampal DOPAC was significantly elevated after 4-wk JP-8 exposure compared to both control groups, suggesting increased dopamine release and metabolism. Findings indicate that behavioral changes do not appear to manifest until wk 3 and 4 of exposure, suggesting the need for longitudinal studies to determine if these behaviors occur due to cumulative exposure, or due to behavioral sensitization related to repeated exposure to aerosolized-vapor JP-8.


Asunto(s)
Aerosoles/toxicidad , Contaminantes Ocupacionales del Aire/toxicidad , Conducta Animal/efectos de los fármacos , Hidrocarburos/toxicidad , Neurotransmisores/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Administración por Inhalación , Análisis de Varianza , Animales , Nivel de Alerta/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Modelos Lineales , Actividad Motora/efectos de los fármacos , Ratas , Ratas Endogámicas F344
17.
Eur J Neurosci ; 24(3): 645-53, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16848797

RESUMEN

Excitatory amino acids such as glutamate play important roles in the central nervous system. We previously demonstrated that a neurosteroid, dehydroepiandrosterone (DHEA), has powerful effects on the cell proliferation of human neural progenitor cells (hNPC) derived from the fetal cortex, and this effect is modulated through NMDA receptor signaling. Here, we show that glutamate can significantly increase the proliferation rates of hNPC. The increased proliferation could be blocked by specific NMDA receptor antagonists, but not other glutamate antagonists for kainate-AMPA or metabotropic receptors. The NR1 subunit of the NMDA receptor was detectable in elongated bipolar or unipolar cells with small cell bodies. These NR1-positive cells were colocalized with GFAP immunoreactivity. Detection of the phosphorylation of cAMP response element-binding protein (pCREB) revealed that a subset of NR1-positive hNPC could respond to glutamate. Furthermore, we hypothesized that glutamate treatment may affect mainly the hNPC with a radial morphology and found that glutamate as well as DHEA selectively affected elongated hNPC; these elongated cells may be a type of radial glial cell. Finally we asked whether the glutamate-responsive hNPC had an increased potential for neurogenesis and found that glutamate-treated hNPC produced significantly more neurons following differentiation. Together these data suggest that glutamate stimulates the division of human progenitor cells with neurogenic potential.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/efectos de los fármacos , Ácido Glutámico/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Células Madre/metabolismo , Diferenciación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Forma de la Célula/fisiología , Células Cultivadas , Sistema Nervioso Central/citología , Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Ácido Glutámico/farmacología , Humanos , Neuroglía/citología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/agonistas , Células Madre/citología , Células Madre/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
18.
Exp Cell Res ; 312(11): 2107-20, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16631163

RESUMEN

Isolation of a true self-renewing stem cell from the human brain would be of great interest as a reliable source of neural tissue. Here, we report that human fetal cortical cells grown in epidermal growth factor expressed low levels of telomerase and telomeres in these cultures shortened over time leading to growth arrest after 30 weeks. Following leukemia inhibitory factor (LIF) supplementation, growth rates and telomerase expression increased. This was best demonstrated following cell cycle synchronization and staining for telomerase using immunocytochemistry. This increase in activity resulted in the maintenance of telomeres at approximately 7 kb for more than 60 weeks in vitro. However, all cultures displayed a lack of oligodendrotye production, decreases in neurogenesis over time and underwent replicative senescence associated with increased expression of p21 before 70 weeks in vitro. Thus, under our culture conditions, these cells are not stable, multipotent, telomerase expressing self-renewing stem cells. They may be more accurately described as human neural progenitor cells (hNPC) with limited lifespan and bi-potent potential (neurons/astrocytes). Interestingly, hNPC follow a course of proliferation, neuronal production and growth arrest similar to that seen during expansion and development of the human cortex, thus providing a possible model neural system. Furthermore, due to their high expansion potential and lack of tumorogenicity, these cells remain a unique and safe source of tissue for clinical transplantation.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula , Senescencia Celular , Corteza Cerebral/citología , Factor de Crecimiento Epidérmico/farmacología , Neuronas/citología , Células Madre/citología , Técnicas de Cultivo de Célula/métodos , Línea Celular , Células Cultivadas , Corteza Cerebral/embriología , Femenino , Humanos , Interleucina-6/farmacología , Factor Inhibidor de Leucemia , Embarazo , Telomerasa/metabolismo
19.
J Neurochem ; 92(3): 462-76, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15659217

RESUMEN

We used human neural stem cells (hNSCs) and their differentiated cultures as a model system to evaluate the mechanism(s) involved in rotenone (RO)- and camptothecin (CA)-induced cytotoxicity. Results from ultrastructural damage and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining indicated that RO-induced cytotoxicity resembled CA-induced apoptosis more than H(2)O(2)-induced necrosis. However, unlike CA-induced, caspase 9/3-dependent apoptosis, there was no increased activity in caspase 9, caspase 3 or poly (ADP-ribose) polymerase (PARP) cleavage in RO-induced cytotoxicity, in spite of time-dependent release of cytochrome c and apoptosis-inducing factor (AIF) following mitochondrial membrane depolarization and a significant increase in reactive oxygen species generation. Equal doses of RO and CA used in hNSCs induced caspase 9/3-dependent apoptosis in differentiated cultures. Time-dependent ATP depletion occurred earlier and to a greater extent in RO-treated hNSCs than in CA-treated hNSCs, or differentiated cultures treated with RO or CA. In conclusion, these results represent a unique ultrastructural and molecular characterization of RO- and CA-induced cytotoxicity in hNSCs and their differentiated cultures. Intracellular ATP levels may play an important role in determining whether neural progenitors or their differentiated cells follow a caspase 9/3-dependent or -independent pathway in response to acute insults from neuronal toxicants.


Asunto(s)
Caspasas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/enzimología , Rotenona/toxicidad , Células Madre/efectos de los fármacos , Células Madre/enzimología , Adenosina Trifosfato/metabolismo , Camptotecina/toxicidad , Caspasa 3 , Caspasa 9 , Muerte Celular/efectos de los fármacos , Diferenciación Celular , Células Cultivadas , Fragmentación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Humanos , Etiquetado Corte-Fin in Situ , Modelos Biológicos , Neuronas/citología , Especies Reactivas de Oxígeno/metabolismo , Células Madre/citología , Inhibidores de Topoisomerasa I , Desacopladores/toxicidad
20.
Proc Natl Acad Sci U S A ; 101(9): 3202-7, 2004 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-14973190

RESUMEN

Dehydroepiandrosterone (DHEA) is a neurosteroid with potential effects on neurogenesis and neuronal survival in humans. However, most studies on DHEA have been performed in rodents, and there is little direct evidence for biological effects on the human nervous system. Furthermore, the mechanism of its action is unknown. Here, we show that DHEA significantly increased the growth rates of human neural stem cells derived from the fetal cortex and grown with both epidermal growth factor (EGF) and leukemia inhibitory factor (LIF). However, it had no effect on cultures grown in either factor alone, suggesting a specific action on the EGF/LIF-responsive cell. Precursors of DHEA such as pregnenolone or six of its major metabolites, had no significant effect on proliferation rates. DHEA did not alter the small number (<3%) of newly formed neuroblasts or the large number (>95%) of nestin-positive precursors. However, the number of glial fibrillary acidic protein-positive cells, its mRNA, and protein were significantly increased by DHEA. We found both N-methyl-d-aspartate and sigma 1 antagonists, but not GABA antagonists, could completely eliminate the effects of DHEA on stem cell proliferation. Finally we asked whether the EGF/LIF/DHEA-responsive stem cells had an increased potential for neurogenesis and found a 29% increase in neuronal production when compared to cultures grown in EGF/LIF alone. Together these data suggest that DHEA is involved in the maintenance and division of human neural stem cells. Given the wide availability of this neurosteroid, this finding has important implications for future use.


Asunto(s)
Corteza Cerebral/embriología , Deshidroepiandrosterona/farmacología , Mitosis/efectos de los fármacos , Neuronas/citología , Células Madre/citología , Secuencia de Bases , Bromodesoxiuridina , Diferenciación Celular , División Celular , Células Cultivadas , Cartilla de ADN , ADN Complementario , Deshidroepiandrosterona/análogos & derivados , Humanos , Índice Mitótico , Neuronas/efectos de los fármacos , Neuropéptidos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Madre/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...