Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Nat Med ; 26(2): 289-299, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31988461

RESUMEN

Young-onset Parkinson's disease (YOPD), defined by onset at <50 years, accounts for approximately 10% of all Parkinson's disease cases and, while some cases are associated with known genetic mutations, most are not. Here induced pluripotent stem cells were generated from control individuals and from patients with YOPD with no known mutations. Following differentiation into cultures containing dopamine neurons, induced pluripotent stem cells from patients with YOPD showed increased accumulation of soluble α-synuclein protein and phosphorylated protein kinase Cα, as well as reduced abundance of lysosomal membrane proteins such as LAMP1. Testing activators of lysosomal function showed that specific phorbol esters, such as PEP005, reduced α-synuclein and phosphorylated protein kinase Cα levels while increasing LAMP1 abundance. Interestingly, the reduction in α-synuclein occurred through proteasomal degradation. PEP005 delivery to mouse striatum also decreased α-synuclein production in vivo. Induced pluripotent stem cell-derived dopaminergic cultures reveal a signature in patients with YOPD who have no known Parkinson's disease-related mutations, suggesting that there might be other genetic contributions to this disorder. This signature was normalized by specific phorbol esters, making them promising therapeutic candidates.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Mutación , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , Adulto , Edad de Inicio , Animales , Diferenciación Celular/genética , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Humanos , Leucocitos Mononucleares/citología , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Fenotipo , Ésteres del Forbol , Fosforilación , Proteómica , Transcriptoma , alfa-Sinucleína/metabolismo
2.
Proc Natl Acad Sci U S A ; 115(4): E762-E771, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29311338

RESUMEN

Increasing evidence suggests that early neurodevelopmental defects in Huntington's disease (HD) patients could contribute to the later adult neurodegenerative phenotype. Here, by using HD-derived induced pluripotent stem cell lines, we report that early telencephalic induction and late neural identity are affected in cortical and striatal populations. We show that a large CAG expansion causes complete failure of the neuro-ectodermal acquisition, while cells carrying shorter CAGs repeats show gross abnormalities in neural rosette formation as well as disrupted cytoarchitecture in cortical organoids. Gene-expression analysis showed that control organoid overlapped with mature human fetal cortical areas, while HD organoids correlated with the immature ventricular zone/subventricular zone. We also report that defects in neuroectoderm and rosette formation could be rescued by molecular and pharmacological approaches leading to a recovery of striatal identity. These results show that mutant huntingtin precludes normal neuronal fate acquisition and highlights a possible connection between mutant huntingtin and abnormal neural development in HD.


Asunto(s)
Enfermedad de Huntington/fisiopatología , Neurogénesis , Línea Celular , Polaridad Celular , Humanos , Enfermedad de Huntington/genética , Células Madre Pluripotentes Inducidas , Telencéfalo/citología
3.
Gene Ther ; 24(4): 245-252, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28276446

RESUMEN

Injecting proteins into the central nervous system that stimulate neuronal growth can lead to beneficial effects in animal models of disease. In particular, glial cell line-derived neurotrophic factor (GDNF) has shown promise in animal and cell models of Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis (ALS). Here, systemic AAV9-GDNF was delivered via tail vein injections to young rats to determine whether this could be a safe and functional strategy to treat the SOD1G93A rat model of ALS and, therefore, translated to a therapy for ALS patients. We found that GDNF administration in this manner resulted in modest functional improvement, whereby grip strength was maintained for longer and the onset of forelimb paralysis was delayed compared to non-treated rats. This did not, however, translate into an extension in survival. In addition, ALS rats receiving GDNF exhibited slower weight gain, reduced activity levels and decreased working memory. Collectively, these results confirm that caution should be applied when applying growth factors such as GDNF systemically to multiple tissues.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Sistema Nervioso Central/fisiopatología , Factor Neurotrófico Derivado de la Línea Celular Glial/uso terapéutico , Neuronas Motoras/patología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Sistema Nervioso Central/efectos de los fármacos , Dependovirus/genética , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Factor Neurotrófico Derivado de la Línea Celular Glial/efectos adversos , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Fuerza de la Mano/fisiología , Humanos , Neuronas Motoras/metabolismo , Ratas , Médula Espinal/efectos de los fármacos , Médula Espinal/fisiopatología , Superóxido Dismutasa/genética
4.
Gene Ther ; 23(5): 424-37, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26863047

RESUMEN

Numerous gene and cell therapy strategies are being developed for the treatment of neurodegenerative disorders. Many of these strategies use constitutive expression of therapeutic transgenic proteins, and although functional in animal models of disease, this method is less likely to provide adequate flexibility for delivering therapy to humans. Ligand-inducible gene expression systems may be more appropriate for these conditions, especially within the central nervous system (CNS). Mifepristone's ability to cross the blood-brain barrier makes it an especially attractive ligand for this purpose. We describe the production of a mifepristone-inducible vector system for regulated expression of transgenes within the CNS. Our inducible system used a lentivirus-based vector platform for the ex vivo production of mifepristone-inducible murine neural progenitor cells that express our transgenes of interest. These cells were processed through a series of selection steps to ensure that the cells exhibited appropriate transgene expression in a dose-dependent and temporally controlled manner with minimal background activity. Inducible cells were then transplanted into the brains of rodents, where they exhibited appropriate mifepristone-inducible expression. These studies detail a strategy for regulated expression in the CNS for use in the development of safe and efficient gene therapy for neurological disorders.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Terapia Genética , Células-Madre Neurales/trasplante , Enfermedades Neurodegenerativas/terapia , Trasplante de Células Madre , Animales , Barrera Hematoencefálica/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/patología , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos , Humanos , Lentivirus/genética , Ratones , Mifepristona/farmacología , Enfermedades Neurodegenerativas/genética , Células Madre , Transgenes/genética
5.
Integr Biol (Camb) ; 4(12): 1522-31, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23147909

RESUMEN

The development of effective stem cell-based therapies for treating brain disorders is keenly dependent upon an understanding of how to generate specific neural cell types and organize them into functional, higher-order tissues analogous to those of the cerebral cortex. Studies of cortical development have revealed that the proper formation of the human cerebral cortex results from specific intercellular interactions and soluble signaling between the highly-proliferative region occupied by dividing neural stem cells and an adjacent region of active neurogenesis and neural migration. However, the factors responsible for establishing this key asymmetrical proliferative-neurogenic architecture are not entirely known. Fibroblast growth factor 2 (FGF-2) is observed in a ventricular-pial gradient during in vivo development and has been previously shown to have effects on both human neural stem cell (hNSC) proliferation and neurogenesis. Here we have adapted a microfluidic approach for creating stable concentration gradients in 3D hydrogels to explore whether FGF-2 gradients can establish defined regions of proliferation and neurogenesis in hNSC cultures. Exponential but not linear FGF-2 gradients between 0-2 ng mL(-1) were able to preferentially boost the percentage of TuJ1(+) neurons in the low concentration regions of the gradient and at levels significantly higher than in non-gradient controls. However, no gradient-dependent localization was observed for dividing hNSCs or hNSC-derived intermediate progenitors. These data suggest that exponential FGF2 gradients are useful for generating asymmetric neuron cultures, but require contributions from other factors to recapitulate the highly-proliferative ventricular zone niche. The relevance of the findings of this study to in vivo cortical development must be more cautiously stated given the artifactual nature of hNSCs and the inability of any in vitro system to fully recapitulate the chemical complexity of the developing cortex. However, it is quite possible that exponential FGF2 gradients are employed in vivo to establish or maintain an asymmetric distribution of neurons in the ventricular-pial axis of the developing cerebral cortex.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Factor 2 de Crecimiento de Fibroblastos/fisiología , Humanos , Hidrogeles , Técnicas Analíticas Microfluídicas , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Proteínas Recombinantes/administración & dosificación , Biología de Sistemas , Ingeniería de Tejidos
6.
Biochim Biophys Acta ; 1792(4): 353-63, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19419698

RESUMEN

Stem cells are central to the development and maintenance of many tissues. This is due to their capacity for extensive proliferation and differentiation into effector cells. More recently it has been shown that the proliferative and differentiative ability of stem cells decreases with age, suggesting that this may play a role in tissue aging. Down syndrome (DS), is associated with many of the signs of premature tissue aging including T-cell deficiency, increased incidence of early Alzheimer-type, Myelodysplastic-type disease and leukaemia. Previously we have shown that both hematopoietic (HSC) and neural stem cells (NSC) in patients affected by DS showed signs of accelerated aging. In this study we tested the hypothesis that changes in gene expression in HSC and NSC of patients affected by DS reflect changes occurring in stem cells with age. The profiles of genes expressed in HSC and NSC from DS patients highlight pathways associated with cellular aging including a downregulation of DNA repair genes and increases in proapoptotic genes, s-phase cell cycle genes, inflammation and angiogenesis genes. Interestingly, Notch signaling was identified as a potential hub, which when deregulated may drive stem cell aging. These data suggests that DS is a valuable model to study early events in stem cell aging.


Asunto(s)
Senescencia Celular , Síndrome de Down/metabolismo , Regulación de la Expresión Génica , Modelos Biológicos , Receptores Notch/metabolismo , Transducción de Señal , Células Madre/metabolismo , Biología de Sistemas , Proteínas Wnt/metabolismo , Anciano , Anciano de 80 o más Años , Preescolar , Síndrome de Down/genética , Síndrome de Down/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Lactante , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Notch/genética , Células Madre/patología , Proteínas Wnt/genética
7.
Gene Ther ; 13(5): 379-88, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16355116

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) has been shown to increase the survival and functioning of dopamine neurons in a variety of animal models and some recent human trials. However, delivery of any protein to the brain remains a challenge due to the blood/brain barrier. Here we show that human neural progenitor cells (hNPC) can be genetically modified to release glycosylated GDNF in vitro under an inducible promoter system. hNPC-GDNF were transplanted into the striatum of rats 10 days following a partial lesion of the dopamine system. At 2 weeks following transplantation, the cells had migrated within the striatum and were releasing physiologically relevant levels of GDNF. This was sufficient to increase host dopamine neuron survival and fiber outgrowth. At 5 weeks following grafting there was a strong trend towards functional improvement in transplanted animals and at 8 weeks the cells had migrated to fill most of the striatum and continued to release GDNF with transport to the substantia nigra. These cells could also survive and release GDNF 3 months following transplantation into the aged monkey brain. No tumors were found in any animal. hNPC can be genetically modified, and thereby represent a safe and powerful option for delivering growth factors to specific targets within the central nervous system for diseases such as Parkinson's.


Asunto(s)
Encéfalo/metabolismo , Terapia Genética/métodos , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Neuronas/fisiología , Trastornos Parkinsonianos/terapia , Trasplante de Células Madre/métodos , Animales , Western Blotting/métodos , Dopamina/metabolismo , Vectores Genéticos/administración & dosificación , Factor Neurotrófico Derivado de la Línea Celular Glial/análisis , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Haplorrinos , Humanos , Inmunohistoquímica/métodos , Lentivirus/genética , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Células Madre/fisiología , Transducción Genética/métodos
8.
J Neurosci Res ; 76(2): 174-83, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15048915

RESUMEN

We have examined the effects of predifferentiation and energy substrate deprivation on long-term expanded human neural precursor cells (HNPCs). The pre-differentiation of HNPC cultures produced large numbers of neurons (>60%) and mature glial cells capable of generating glycogen stores that protected the neuronal population from experimental metabolic stress. When predifferentiated HNPCs were transplanted into intact adult rat hippocampus, fewer cells survived compared to undifferentiated HNPC transplants. This cell death was completely attenuated, however, when predifferentiated HNPC cultures were pretreated to boost glial energy stores and resulted in greatly increased neuronal survival in vivo. The transplanted cells primarily engrafted within the granular layer of the dentate gyrus, where a large proportion of the predifferentiated HNPCs co-expressed neuronal markers whereas most HNPCs outside of the neuronal layer did not, indicating that the predifferentiated cells remained capable of responding to local cues in the adult brain. Undifferentiated HNPCs migrated more widely in the brain after grafting than did the predifferentiated cells, which generally remained within the hippocampus.


Asunto(s)
Supervivencia Celular/fisiología , Sistema Nervioso Central/citología , Neuronas/fisiología , Trasplante de Células Madre/métodos , Células Madre/fisiología , Análisis de Varianza , Animales , Recuento de Células/métodos , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Sistema Nervioso Central/metabolismo , Embrión de Mamíferos , Proteína Ácida Fibrilar de la Glía/metabolismo , Glucosa/administración & dosificación , Glucógeno/metabolismo , Humanos , Imagenología Tridimensional/métodos , Inmunohistoquímica/métodos , Técnicas In Vitro , L-Lactato Deshidrogenasa/metabolismo , Microscopía Confocal/métodos , Fosfopiruvato Hidratasa/metabolismo , Ratas , Factores de Tiempo , Tubulina (Proteína)/metabolismo
9.
Genes Brain Behav ; 2(3): 179-86, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12931791

RESUMEN

The clinical characteristics of Down's syndrome (DS), or trisomy 21, are caused by errors that occur during development. In addition to mental retardation, DS individuals have craniofacial abnormalities, clinical defects of the heart, gut and immune system, as well as predisposition to certain diseases, such as leukemias and Alzheimer's disease. To explain the developmental mechanisms that cause these traits, it is necessary to look at how developmental processes in DS compare to normal development. The neurological characteristics of DS are established during the prenatal and early postnatal period in humans, when the bulk of brain development occurs. Mouse models of DS have provided a useful way of studying DS neural development. However, there are clearly significant differences between rodent and human biology that may not be reflected in mouse models. Recent advances in stem cell biology now allow the generation of human neural tissue in the culture dish (Ostenfeld & Svendsen 2003). Stem cells offer a novel model system to study alterations in neuron development in developmental disorders such as DS.


Asunto(s)
Corteza Cerebral/patología , Síndrome de Down/patología , Neuronas/patología , Células Madre/patología , Animales , Corteza Cerebral/crecimiento & desarrollo , Modelos Animales de Enfermedad , Síndrome de Down/genética , Humanos , Ratones
10.
Adv Tech Stand Neurosurg ; 28: 3-89, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12627808

RESUMEN

1. Neural stem cells can be cultured from the CNS of different mammalian species at many stages of development. They have an extensive capacity for self-renewal and will proliferate ex vivo in response to mitogenic growth factors or following genetic modification with immortalising oncogenes. Neural stem cells are multipotent since their differentiating progeny will give rise to the principal cellular phenotypes comprising the mature CNS: neurons, astrocytes and oligodendrocytes. 2. Neural stem cells can also be derived from more primitive embryonic stem (ES) cells cultured from the blastocyst. ES cells are considered to be pluripotent since they can give rise to the full cellular spectrum and will, therefore, contribute to all three of the embryonic germ layers: endoderm, mesoderm and ectoderm. However, pluripotent cells have also been derived from germ cells and teratocarcinomas (embryonal carcinomas) and their progeny may also give rise to the multiple cellular phenotypes contributing to the CNS. In a recent development, ES cells have also been isolated and grown from human blastocysts, thus raising the possibility of growing autologous stem cells when combined with nuclear transfer technology. 3. There is now an emerging recognition that the adult mammalian brain, including that of primates and humans, harbours stem cell populations suggesting the existence of a previously unrecognised neural plasticity to the mature CNS, and thereby raising the possibility of promoting endogenous neural reconstruction. 4. Such reports have fuelled expectations for the clinical exploitation of neural stem cells in cell replacement or recruitment strategies for the treatment of a variety of human neurological conditions including Parkinson's disease (PD), Huntington's disease, multiple sclerosis and ischaemic brain injury. Owing to their migratory capacity within the CNS, neural stem cells may also find potential clinical application as cellular vectors for widespread gene delivery and the expression of therapeutic proteins. In this regard, they may be eminently suitable for the correction of genetically-determined CNS disorders and in the management of certain tumors responsive to cytokines. Since large numbers of stem cells can be generated efficiently in culture, they may obviate some of the technical and ethical limitations associated with the use of fresh (primary) embryonic neural tissue in current transplantation strategies. 5. While considerable recent progress has been made in terms of developing new techniques allowing for the long-term culture of human stem cells, the successful clinical application of these cells is presently limited by our understanding of both (i) the intrinsic and extrinsic regulators of stem cell proliferation and (ii) those factors controlling cell lineage determination and differentiation. Although such cells may also provide accessible model systems for studying neural development, progress in the field has been further limited by the lack of suitable markers needed for the identification and selection of cells within proliferating heterogeneous populations of precursor cells. There is a further need to distinguish between the committed fate (defined during normal development) and the potential specification (implying flexibility of fate through manipulation of its environment) of stem cells undergoing differentiation. 6. With these challenges lying ahead, it is the opinion of the authors that stem-cell therapy is likely to remain within the experimental arena for the foreseeable future. In this regard, few (if any) of the in vivo studies employing neural stem cell grafts have shown convincingly that behavioural recovery can be achieved in the various model paradigms. Moreover, issues relating to the quality control of cultured cells and their safety following transplantation have only begun to be addressed. 7. While on the one hand cell biotechnologists have been quick to realise the potential commercial value, human stem cell research and its clinical applications has been the subject of intense ethical and legislative considerations. The present chapter aims to review some recent aspects of stem cell research applicable to developmental neurobiology and the potential applications in clinical neuroscience.


Asunto(s)
Neurobiología/tendencias , Células Madre/fisiología , Animales , Humanos
11.
Brain Res Dev Brain Res ; 134(1-2): 43-55, 2002 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-11947936

RESUMEN

Neural precursor cells were isolated from various regions of the developing rat and human brain and grown in culture as aggregates termed neurospheres. We asked whether cells within human and rodent neurospheres are identical, or whether they have species specific characteristics or differences based on their region of origin. Under our culture conditions, rodent neurospheres isolated from the cortex (ctxNS) and striatum (strNS) grew faster than those from the mesencephalon (mesNS), but stopped growing after only eight to ten population doublings. In contrast, human neurospheres under identical culture conditions, continued to grow for over 40 population doublings. Following migration and differentiation of both rodent and human cultures, ctxNS and strNS generated high numbers of small neurons whereas mesNS generated small numbers of large neurons with many long fibres. Only very rare neurons from mesNS expressed dopaminergic markers, and thus may require further signals to fully mature. While the rat neurospheres generated high numbers of oligodendrocytes, very few were found to develop from human neurospheres from any region after a few weeks of passaging. FACS analysis revealed a unique population of smaller cells within human strNS and ctxNS, which appeared to be neuronal progenitors. However, large cells within neurospheres were capable of generating these small neuronal progenitors following further proliferation. Together, our data show that rat and human neurospheres have unique characteristics with regard to growth and differentiation, and that the majority of precursor cells within neurospheres are regionally specified to generate set numbers of neurons. These findings have important implications for understanding the nature of proliferating neural precursors isolated from the developing CNS, and their potential for brain repair.


Asunto(s)
Encéfalo/citología , Encéfalo/fisiología , Neuronas/citología , Neuronas/fisiología , Ratas/fisiología , Esferoides Celulares/citología , Esferoides Celulares/fisiología , Animales , Diferenciación Celular , División Celular/fisiología , Movimiento Celular , Tamaño de la Célula , Células Cultivadas , Corteza Cerebral/citología , Cuerpo Estriado/citología , Humanos , Mesencéfalo/citología , Oligodendroglía/citología , Especificidad de la Especie , Factores de Tiempo
12.
Gene Ther ; 9(4): 245-55, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11896463

RESUMEN

Human neural progenitor cells (hNPCs) represent an attractive source for cell therapy of neurological disorders. Genetic modification of hNPCs may allow a controlled release of therapeutic proteins, suppress immune rejection, or produce essential neurotransmitters. In search of an effective gene delivery vehicle, we evaluated the efficiency of a recombinant adeno-associated viral (rAAV) vector expressing enhanced green fluorescent protein (CAGegfp). Our study demonstrated that CAGegfp efficiently transduced both proliferating and differentiated hNPCs in vitro. EGFP expression was detected as early as 1 day after exposure to CAGegfp and was detectable for up to 4 months. Following transduction, the growth rate of hNPCs slowed down, but they were still able to differentiate into neurons and glia. Furthermore, CAGegfp-modified hNPCs survived, differentiated and expressed EGFP after transplanting into spinal cord of adult rats. Our results indicated that rAAV vectors might be a useful tool in hNPC-based cell and gene therapy for neurological disorders.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos , Neuronas/trasplante , Trasplante de Células Madre , Transducción Genética/métodos , Animales , Diferenciación Celular , División Celular , Embrión de Mamíferos/citología , Genes Reporteros , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/genética , Masculino , Neuroglía/citología , Neuronas/citología , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología , Células Madre/citología
14.
J Mol Neurosci ; 17(1): 25-33, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11665860

RESUMEN

Sequential exposure to members of the neurotrophin family, nerve growth factor (NGF), neurotrophin-type 3 (NT3), brain-derived neurotrophic factor (BDNF), and neurotrophin-type 4 (NT4), determines the generation, survival, and maturation of developing neurons. The effects of neurotrophins depend on the stage of development and the target cell population. However, the nature of the responding cells is often unclear. In this study neurotrophin responsiveness was analyzed in murine embryonic striatal precursors and neurons. Individual neurotrophin-responsive cells were identified based on activation of intracellular signaling pathways to the transcription factor CREB and were further characterized using differentiation-stage specific markers. A dramatic developmentally regulated decrease in BDNF responsiveness was observed: BDNF targeted more than 40% of striatal neurons at E14 but only 12% at E18. The percentage of NT3-responsive neurons also moderately decreased during development while no change was observed in the fraction of neuronal cells targeted by NT4 and NGF. A different type of developmental change was found in striatal precursors. BDNF, NT3, and NT4 each targeted about 15% of striatal precursors at E14 but no NGF responsive-precursors were detected at this age. In contrast, only NT3 and NGF could induce a response in precursor cells at E18. NGF-responsive precursors shared a distinct morphology with a large cell body and high levels of nestin expression. These results indicate that during striatal development, the regulation of neurotrophin responsiveness is different in neurons and precursor cells.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Cuerpo Estriado/embriología , Factor de Crecimiento Nervioso/farmacología , Factores de Crecimiento Nervioso/farmacología , Neuronas/efectos de los fármacos , Neurotrofina 3/farmacología , Animales , Tamaño de la Célula , Células Cultivadas , Cuerpo Estriado/citología , Cuerpo Estriado/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Inmunohistoquímica , Ratones , Neuronas/citología , Neuronas/fisiología , Fármacos Neuroprotectores/farmacología
15.
Nat Biotechnol ; 19(5): 475-9, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11329020

RESUMEN

Cells isolated from the embryonic, neonatal, and adult rodent central nervous system divide in response to epidermal growth factor (EGF) and fibroblast growth factor 2 (FGF-2), while retaining the ability to differentiate into neurons and glia. These cultures can be grown in aggregates termed neurospheres, which contain a heterogeneous mix of both multipotent stem cells and more restricted progenitor populations. Neurospheres can also be generated from the embryonic human brain and in some cases have been expanded for extended periods of time in culture. However, the mechanisms controlling the number of neurons generated from human neurospheres are poorly understood. Here we show that maintaining cell-cell contact during the differentiation stage, in combination with growth factor administration, can increase the number of neurons generated under serum-free conditions from 8% to > 60%. Neurotrophic factors 3 and 4 (NT3, NT4) and platelet-derived growth factor (PDGF) were the most potent, and acted by increasing neuronal survival rather than inducing neuronal phenotype. Following differentiation, the neurons could survive dissociation and either replating or transplantation into the adult rat brain. This experimental system provides a practically limitless supply of enriched, non-genetically transformed neurons. These should be useful for both neuroactive drug screening in vitro and possibly cell therapy for neurodegenerative diseases.


Asunto(s)
División Celular/efectos de los fármacos , Sustancias de Crecimiento/farmacología , Neuronas/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Encéfalo/citología , Encéfalo/embriología , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Trasplante de Células/métodos , Células Cultivadas , Factor 2 de Crecimiento de Fibroblastos/farmacología , Sustancias de Crecimiento/fisiología , Humanos , Factores de Crecimiento Nervioso/farmacología , Neuronas/citología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas , Células Madre/citología
16.
Trends Mol Med ; 7(4): 144-6, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11286926

RESUMEN

There is growing excitement over stem cell biology. It has stirred strong ethical and moral debates over the status and rights of small clusters of cells. It has promised a panacea for illnesses ranging from diabetes to stroke. It has challenged historical dogmas in developmental biology. There have been many commentaries on all of these issues in prominent journals and newspapers over recent months. In this article, we take a critical look at new data that underpin the last of these claims: the chimeric stem cell.


Asunto(s)
Quimera , Células Madre/citología , Células Madre/metabolismo , Animales , Astrocitos/citología , Células de la Médula Ósea/citología , Diferenciación Celular , Sistema Nervioso Central/citología , Humanos , Ratones , Modelos Biológicos , Neuronas/citología , Fenotipo
17.
J Neuroimmunol ; 112(1-2): 35-46, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11108931

RESUMEN

Neural stem cells are currently considered very hopeful candidates for cell replacement therapy in neurodegenerative pathologies such as Parkinson's disease. Here we show that different cell types derived from neurospheres amplified in vitro can be identified by FACS analysis relying solely on physical parameters (FSC/SSC) or autofluorescence. Additionally, after treatment with a panel of inflammatory cytokines, neurospheres and their differentiated progeny were shown to express MHC antigens which could potentially cause transplant rejection. Astrocytes expressed the highest levels of MHC. Hence removing such cells prior to transplantation could potentially optimise transplant survival.


Asunto(s)
Citometría de Flujo , Antígenos de Histocompatibilidad Clase II/análisis , Antígenos de Histocompatibilidad Clase I/análisis , Neuronas/fisiología , Células Madre/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Interferones/farmacología , Ratas , Ratas Endogámicas Lew , Factor de Necrosis Tumoral alfa/farmacología
19.
Exp Neurol ; 164(1): 215-26, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10877932

RESUMEN

Worldwideattention is presently focused on proliferating populations of neural precursor cells as an in vitro source of tissue for neural transplantation and brain repair. However, successful neuroreconstruction is contingent upon their capacity to integrate within the host CNS and the absence of tumorigenesis. Here we show that human neural precursor cells express very low levels of telomerase at early passages (less than 20 population doublings), but that this decreases to undetectable levels at later passages. In contrast, rodent neural precursors express high levels of telomerase at both early and late passages. The human neural precursors also have telomeres (approximately 12 kbp) significantly shorter than those of their rodent counterparts (approximately 40 kbp). Human neural precursors were then expanded 100-fold prior to intrastriatal transplantation in a rodent model of Parkinson's disease. To establish the effects of implanted cell number on survival and integration, precursors were transplanted at either 200,000, 1 million, or 2 million cells per animal. Interestingly, the smaller transplants were more likely to extend neuronal fibers and less likely to provoke immune rejection than the largest transplants in this xenograft model. Cellular proliferation continued immediately post-transplantation, but by 20 weeks there were virtually no dividing cells within any of the grafts. In contrast, fiber outgrowth increased gradually over time and often occupied the entire striatum at 20 weeks postgrafting. Transient expression of tyrosine hydroxylase-positive cells within the grafts was found in some animals, but this was not sustained at 20 weeks and had no functional effects. For Parkinson's disease, the principal aim now is to induce the dopaminergic phenotype in these cells prior to transplantation. However, given the relative safety profile for these human cells and their capacity to extend fibers into the adult rodent brain, they may provide the ideal basis for the repair of other lesions of the CNS where extensive axonal outgrowth is required.


Asunto(s)
Neuronas/citología , Neuronas/enzimología , Células Madre/enzimología , Telomerasa/biosíntesis , Animales , Axones/metabolismo , Axones/ultraestructura , Trasplante de Tejido Encefálico , Recuento de Células , Diferenciación Celular , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Femenino , Trasplante de Tejido Fetal , Factor 2 de Crecimiento de Fibroblastos/farmacología , Supervivencia de Injerto , Humanos , Fibras Nerviosas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/trasplante , Oxidopamina , Ratas , Trasplante de Células Madre , Células Madre/citología , Células Madre/efectos de los fármacos , Telómero/ultraestructura , Tiempo , Trasplante Heterólogo , Tubulina (Proteína)/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
20.
Cell Transplant ; 9(2): 139-52, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10811389

RESUMEN

A large number of crippling neurological conditions result from the loss of certain cell populations from the nervous system through disease or injury, and these cells are not intrinsically replaced. Mounting evidence now suggests that replacement of depleted cell populations by transplantation may be of functional benefit in many such diseases. A diverse range of cell populations is vulnerable, and the loss of specific populations results in circumscribed deficits in different conditions. This diversity presents a considerable challenge if cell replacement therapy is to become widely applicable in the clinical domain, because each condition has specific requirements for the phenotype, developmental stage, and number of cells required. An ideal cell for universal application in cell replacement therapy would possess several key properties: it would be highly proliferative, allowing the ex vivo production of large numbers of cells from minimal donor material; it would also remain immature and phenotypically plastic such that it could differentiate into appropriate neural and glial cell types on, or prior to, transplantation. Critically, both proliferation and differentiation would be controllable. This review considers some of the evidence that stem cells exist in the central nervous system and that they may possess characteristics that make them ideal for broad application in cell replacement therapy.


Asunto(s)
Trasplante de Tejido Encefálico , Trasplante de Células , Trasplante de Células Madre , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Sistema Nervioso Central/citología , Humanos , Enfermedades del Sistema Nervioso/terapia , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...