Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Intern Med ; 276(1): 25-40, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24708237

RESUMEN

High-grade gliomas remain incurable and lethal. Through the availability of the stem-like cells responsible for glioblastoma (GB) formation, expansion, resilience and recurrence, the discovery of glioma cancer stem cells (GCSCs) is revolutionizing this field. GCSCs provide an unprecedented opportunity to reproduce and study GB pathophysiology more accurately. This critically emphasizes our ability to unambiguously identify, isolate and investigate cells that do qualify as GCSCs, to use them as a potential model that is truly predictive of GBs and of their regulation and response to therapeutic agents. We review this concept against the background of key findings on somatic, neural and solid tumour stem cells (SCs), also taking into account the emerging phenomenon of phenotypic SC plasticity. We suggest that basic approaches in these areas can be imported into the GCSC field, so that the same functional method used to identify normal somatic SCs becomes the most appropriate to define GCSCs. This, combined with knowledge of the cellular and molecular basis of normal adult neurogenesis, promises to improve the identification of GCSCs and of selective markers, as well as the development of innovative, more specific and efficacious antiglioma strategies.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Células Madre Neoplásicas/citología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones SCID , Células Madre Neoplásicas/patología , Fenotipo , Nicho de Células Madre/fisiología
2.
Stem Cell Res ; 77: 103412, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38613988

RESUMEN

Genetic expansions of the hexanucleotide repeats (GGGGCC) in the C9orf72 gene appear in approximately 40% of patients with familial ALS and 7% of patients with sporadic ALS in the European population, making this mutation one of the most prevalent genetic mutations in ALS. Here, we generated a human induced pluripotent stem cell (hiPSC) line from the dermal fibroblasts of a patient carrying a 56-repeat expansion in an ALS disease-causing allele of C9orf72. These iPSCs showed stable amplification in vitro with normal karyotype and high expression of pluripotent markers and differentiated spontaneously in vivo into three germ layers.


Asunto(s)
Esclerosis Amiotrófica Lateral , Proteína C9orf72 , Expansión de las Repeticiones de ADN , Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Diferenciación Celular , Fibroblastos/metabolismo , Línea Celular , Masculino
3.
Cell Death Dis ; 15(8): 639, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39217148

RESUMEN

Pre-clinical trials have demonstrated the neuroprotective effects of transplanted human neural stem cells (hNSCs) during the post-ischemic phase. However, the exact neuroprotective mechanism remains unclear. Tunneling nanotubes (TNTs) are long plasma membrane bridges that physically connect distant cells, enabling the intercellular transfer of mitochondria and contributing to post-ischemic repair processes. Whether hNSCs communicate through TNTs and their role in post-ischemic neuroprotection remains unknown. In this study, non-immortalized hNSC lines derived from fetal human brain tissues were examined to explore these possibilities and assess the post-ischemic neuroprotection potential of these hNSCs. Using Tau-STED super-resolution confocal microscopy, live cell time-lapse fluorescence microscopy, electron microscopy, and direct or non-contact homotypic co-cultures, we demonstrated that hNSCs generate nestin-positive TNTs in both 3D neurospheres and 2D cultures, through which they transfer functional mitochondria. Co-culturing hNSCs with differentiated SH-SY5Y (dSH-SY5Y) revealed heterotypic TNTs allowing mitochondrial transfer from hNSCs to dSH-SY5Y. To investigate the role of heterotypic TNTs in post-ischemic neuroprotection, dSH-SY5Y were subjected to oxygen-glucose deprivation (OGD) followed by reoxygenation (OGD/R) with or without hNSCs in direct or non-contact co-cultures. Compared to normoxia, OGD/R dSH-SY5Y became apoptotic with impaired electrical activity. When OGD/R dSH-SY5Y were co-cultured in direct contact with hNSCs, heterotypic TNTs enabled the transfer of functional mitochondria from hNSCs to OGD/R dSH-SY5Y, rescuing them from apoptosis and restoring the bioelectrical profile toward normoxic dSH-SY5Y. This complete neuroprotection did not occur in the non-contact co-culture. In summary, our data reveal the presence of a functional TNTs network containing nestin within hNSCs, demonstrate the involvement of TNTs in post-ischemic neuroprotection mediated by hNSCs, and highlight the strong efficacy of our hNSC lines in post-ischemic neuroprotection. Human neural stem cells (hNSCs) communicate with each other and rescue ischemic neurons through nestin-positive tunneling nanotubes (TNTs). A Functional mitochondria are exchanged via TNTs between hNSCs. B hNSCs transfer functional mitochondria to ischemic neurons through TNTs, rescuing neurons from ischemia/reperfusion ROS-dependent apoptosis.


Asunto(s)
Comunicación Celular , Técnicas de Cocultivo , Mitocondrias , Células-Madre Neurales , Neuronas , Humanos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Neuronas/metabolismo , Mitocondrias/metabolismo , Encéfalo/metabolismo , Encéfalo/embriología , Diferenciación Celular , Nanotubos/química , Feto , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Estructuras de la Membrana Celular
4.
Front Cell Dev Biol ; 11: 1221671, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37886397

RESUMEN

Tunneling nanotubes (TNTs) are long F-actin-positive plasma membrane bridges connecting distant cells, allowing the intercellular transfer of cellular cargoes, and are found to be involved in glioblastoma (GBM) intercellular crosstalk. Glial fibrillary acid protein (GFAP) is a key intermediate filament protein of glial cells involved in cytoskeleton remodeling and linked to GBM progression. Whether GFAP plays a role in TNT structure and function in GBM is unknown. Here, analyzing F-actin and GFAP localization by laser-scan confocal microscopy followed by 3D reconstruction (3D-LSCM) and mitochondria dynamic by live-cell time-lapse fluorescence microscopy, we show the presence of GFAP in TNTs containing functional mitochondria connecting distant human GBM cells. Taking advantage of super-resolution 3D-LSCM, we show the presence of GFAP-positive TNT-like structures in resected human GBM as well. Using H2O2 or the pro-apoptotic toxin staurosporine (STS), we show that GFAP-positive TNTs strongly increase during oxidative stress and apoptosis in the GBM cell line. Culturing GBM cells with STS-treated GBM cells, we show that STS triggers the formation of GFAP-positive TNTs between them. Finally, we provide evidence that mitochondria co-localize with GFAP at the tip of close-ended GFAP-positive TNTs and inside receiving STS-GBM cells. Summarizing, here we found that GFAP is a structural component of TNTs generated by GBM cells, that GFAP-positive TNTs are upregulated in response to oxidative stress and pro-apoptotic stress, and that GFAP interacts with mitochondria during the intercellular transfer. These findings contribute to elucidate the molecular structure of TNTs generated by GBM cells, highlighting the structural role of GFAP in TNTs and suggesting a functional role of this intermediate filament component in the intercellular mitochondria transfer between GBM cells in response to pro-apoptotic stimuli.

5.
Nature ; 444(7120): 761-5, 2006 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-17151667

RESUMEN

Transformed, oncogenic precursors, possessing both defining neural-stem-cell properties and the ability to initiate intracerebral tumours, have been identified in human brain cancers. Here we report that bone morphogenetic proteins (BMPs), amongst which BMP4 elicits the strongest effect, trigger a significant reduction in the stem-like, tumour-initiating precursors of human glioblastomas (GBMs). Transient in vitro exposure to BMP4 abolishes the capacity of transplanted GBM cells to establish intracerebral GBMs. Most importantly, in vivo delivery of BMP4 effectively blocks the tumour growth and associated mortality that occur in 100% of mice after intracerebral grafting of human GBM cells. We demonstrate that BMPs activate their cognate receptors (BMPRs) and trigger the Smad signalling cascade in cells isolated from human glioblastomas (GBMs). This is followed by a reduction in proliferation, and increased expression of markers of neural differentiation, with no effect on cell viability. The concomitant reduction in clonogenic ability, in the size of the CD133+ population and in the growth kinetics of GBM cells indicates that BMP4 reduces the tumour-initiating cell pool of GBMs. These findings show that the BMP-BMPR signalling system--which controls the activity of normal brain stem cells--may also act as a key inhibitory regulator of tumour-initiating, stem-like cells from GBMs and the results also identify BMP4 as a novel, non-cytotoxic therapeutic effector, which may be used to prevent growth and recurrence of GBMs in humans.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Proteína Morfogenética Ósea 4 , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Glioblastoma/metabolismo , Glioblastoma/patología , Glicoproteínas/metabolismo , Humanos , Ratones , Células Madre Neoplásicas/citología , Péptidos/metabolismo , Transducción de Señal/efectos de los fármacos , Trasplante de Células Madre
6.
Science ; 283(5401): 534-7, 1999 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-9915700

RESUMEN

Stem cells are found in various organs where they participate in tissue homeostasis by replacing differentiated cells lost to physiological turnover or injury. An investigation was performed to determine whether stem cells are restricted to produce specific cell types, namely, those from the tissue in which they reside. After transplantation into irradiated hosts, genetically labeled neural stem cells were found to produce a variety of blood cell types including myeloid and lymphoid cells as well as early hematopoietic cells. Thus, neural stem cells appear to have a wider differentiation potential than previously thought.


Asunto(s)
Células Sanguíneas/citología , Células Madre Hematopoyéticas/citología , Prosencéfalo/citología , Células Madre/citología , Animales , Células Sanguíneas/inmunología , Células de la Médula Ósea/inmunología , Diferenciación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Femenino , Antígenos H-2/análisis , Hematopoyesis , Operón Lac , Linfocitos/citología , Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Prosencéfalo/embriología , Bazo/citología , Trasplante de Células Madre , Células Madre/inmunología
7.
Neuron ; 11(5): 951-66, 1993 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8240816

RESUMEN

In cultures of embryonic and adult mouse striatum, we previously demonstrated that EGF induces the proliferation of putative stem cells, which give rise to spheres of undifferentiated cells that can generate neurons and astrocytes. We report here that the spheres of undifferentiated cells contain mRNA and protein for the FGF receptor (FGFR1). Indirect immunocytochemistry demonstrated that many of the cells within the EGF-generated spheres were immunoreactive for FGFR1. Exogenous application of bFGF to the EGF-generated cells induced the proliferation of two progenitor cell types. The first, a bipotent progenitor cell, gave rise to cells with the antigenic and morphological properties of neurons and astrocytes; the other gave rise to cells with neuronal characteristics only. bFGF-generated cells with neuronal morphology exhibited electrophysiological properties indicative of immature central neurons. These results support the hypothesis that sequential actions of growth factors play a role in regulating the generation of neurons and astrocytes in the developing CNS.


Asunto(s)
Astrocitos/citología , Encéfalo/citología , Factor de Crecimiento Epidérmico/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Neuronas/citología , Células Madre/citología , Animales , Astrocitos/efectos de los fármacos , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Electrofisiología , Neuronas/efectos de los fármacos , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/genética , Células Madre/metabolismo , Células Madre/fisiología
8.
Nat Neurosci ; 3(10): 986-91, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11017170

RESUMEN

Distinct cell lineages established early in development are usually maintained throughout adulthood. Thus, adult stem cells have been thought to generate differentiated cells specific to the tissue in which they reside. This view has been challenged; for example, neural stem cells can generate cells that normally originate from a different germ layer. Here we show that acutely isolated and clonally derived neural stem cells from mice and humans could produce skeletal myotubes in vitro and in vivo, the latter following transplantation into adult animals. Myogenic conversion in vitro required direct exposure to myoblasts, and was blocked if neural cells were clustered. Thus, a community effect between neural cells may override such myogenic induction. We conclude that neural stem cells, which generate neurons, glia and blood cells, can also produce skeletal muscle cells, and can undergo various patterns of differentiation depending on exposure to appropriate epigenetic signals in mature tissues.


Asunto(s)
Trasplante de Tejido Encefálico/métodos , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Sistema Nervioso Central/embriología , Músculo Esquelético/embriología , Trasplante de Células Madre , Células Madre/metabolismo , Factores de Edad , Animales , Comunicación Celular/fisiología , Técnicas de Cultivo de Célula/métodos , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/ultraestructura , Técnicas de Cocultivo , Humanos , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Células Madre/ultraestructura
9.
Trends Neurosci ; 19(9): 387-93, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8873356

RESUMEN

Neural precursor cells have been of interest historically as the building blocks of the embryonic CNS and, most recently, as substrates for restorative neurological approaches. The majority of previous in vitro studies of the regulation of neural-cell proliferation by polypeptide growth factors, and in vivo studies of neural lineage, argue for the presence of precursors with limited proliferative or lineage potential in the mammalian CNS. This is in contrast to renewable tissues, such as the blood or immune system, skin epithelium and epithelium of the small intestinal crypts, which contain specialized, self-renewing cells known as stem cells. However, recent in vitro and in vivo studies from our and other laboratories lead us to conclude that neural stem cells, with self-renewal and multilineage potential, are present in the embryonic through to adult mammalian forebrain.


Asunto(s)
Factor de Crecimiento Epidérmico/fisiología , Prosencéfalo/citología , Células Madre/fisiología , Animales , Modelos Biológicos , Prosencéfalo/crecimiento & desarrollo
10.
Brain Pathol ; 9(3): 569-98, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10416994

RESUMEN

The study of the basic physiology of the neural precursors generated during brain development is driven by two inextricably linked goals. First, such knowledge is instrumental to our understanding of how the high degree of cellular complexity of the mature central nervous system (CNS) is generated, and how to dissect the steps of proliferation, fate commitment, and differentiation that lead early pluripotent neural progenitors to give rise to mature CNS cells. Second, it is hoped that the isolation, propagation, and manipulation of brain precursors and, particularly, of multipotent neural stem cells (NSCs), will lead to therapeutic applications in neurological disorders. The debate is still open concerning the most appropriate definition of a stem cell and on how it is best identified, characterized, and manipulated. By adopting an operational definition of NSCs, we review some of the basic findings in this area and elaborate on their potential therapeutic applications. Further, we discuss recent evidence from our two groups that describe, based on that rigorous definition, the isolation and propagation of clones of NSCs from the human fetal brain and illustrate how they have begun to show promise for neural cell replacement and molecular support therapy in models of degenerative CNS diseases. The extensive propagation and engraftment potential of human CNS stem cells may, in the not-too-distant-future, be directed towards genuine clinical therapeutic ends, and may open novel and multifaceted strategies for redressing a variety of heretofore untreatable CNS dysfunctions.


Asunto(s)
Células Clonales/citología , Plasticidad Neuronal/fisiología , Neuronas/citología , Células Madre/citología , Animales , Encéfalo/citología , Trasplante de Tejido Encefálico/métodos , Trasplante de Tejido Encefálico/tendencias , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular , Linaje de la Célula , Separación Celular , Células Clonales/fisiología , Células Clonales/trasplante , Embrión de Mamíferos , Feto , Humanos , Ratones , Ratas , Trasplante de Células Madre , Células Madre/fisiología
11.
Neuroscience ; 72(2): 467-75, 1996 May.
Artículo en Inglés | MEDLINE | ID: mdl-8737416

RESUMEN

A novel in vitro system which allows extensive culturing of multipotential stem cells from mouse brain has made it possible to test whether enzymes that metabolize androgens and progestagens are present in undifferentiated central nervous system progenitor cells. Embryonic day 14 striatal cells were grown in the presence of either 20 ng/ml of epidermal growth factor (which prevents cell differentiation), or 2% fetal bovine serum (facilitating differentiation). Differentiation was complete by 35 days in vitro when the cell population comprised 86 +/- 2.0% astrocytes, 6 +/- 0.7% neurons 1.6 +/- 0.5% oligodendrocytes and 6.4 +/- 0.5% undifferentiated cells. No changes in the proportions of cell type were observed thereafter (38 and 45 days in vitro). 5 alpha-Reductive conversion (by 5 alpha-reductase) of testosterone and progesterone into dihydrotestosterone and dihydroprogesterone, and subsequent 3-alpha hydroxylation (by 3 alpha-hydroxysteroid dehydrogenase) to 3 alpha-diol and tetrahydroprogesterone were assayed in the cultures at 35, 38 and 45 days in vitro. Undifferentiated epidermal growth factor-treated cells (controls) formed about 10 times more dihydroprogesterone than dihydrotestosterone. Conversions of dihydrotestosterone and dihydroprogesterone, respectively, into 3 alpha-diol and tetrahydroprogesterone were very similar. In the fetal bovine serum-treated differentiating cells, 5 alpha-reductase converting progesterone increased at 38 days in vitro, and remained similarly elevated at 42 days in vitro (4 times). However, the conversion of testosterone into dihydrotestosterone remained at control levels up to 42 days in vitro when an increase was observed. 3 alpha-Hydroxysteroid dehydrogenase activity converting dihydroprogesterone but not dihydrotestosterone was increased at 38 and 42 days in vitro. These results show that undifferentiated central nervous system cells possess androgen and progestagen metabolizing enzymes which are strongly influenced by the cellular differentiation/maturation process.


Asunto(s)
Sistema Nervioso Central/citología , Sistema Nervioso Central/enzimología , Células Madre/enzimología , Esteroides/metabolismo , 20-alfa-Dihidroprogesterona/farmacología , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , Andrógenos/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Sistema Nervioso Central/efectos de los fármacos , Femenino , Técnica del Anticuerpo Fluorescente Directa , Cinética , Ratones , Neostriado/citología , Neostriado/efectos de los fármacos , Neostriado/crecimiento & desarrollo , Neuroglía/efectos de los fármacos , Neuroglía/enzimología , Neuroglía/ultraestructura , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/ultraestructura , Embarazo , Progestinas/farmacología , Células Madre/efectos de los fármacos
12.
J Neurotrauma ; 16(8): 689-93, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10511241

RESUMEN

In this work, we show that the embryonic human brain contains multipotent central nervous system (CNS) stem cells, which may provide a continuous, standardized source of human neurons that could virtually eliminate the use of primary human fetal brain tissue for intracerebral transplantation. Multipotential stem cells can be isolated from the developing human CNS in a reproducible fashion and can be exponentially expanded for longer than 2 years. This allows for the establishment of continuous, nontransformed neural cell lines, which can be frozen and banked. By clonal analysis, reverse transcription polymerase chain reaction, and electrophysiological assay, we found that over such long-term culturing these cells retain both multipotentiality and an unchanged capacity for the generation of neuronal cells, and that they can be induced to differentiate into catechlaminergic neurons. Finally, when transplanted into the brain of adult rodents immunosuppressed by cyclosporin A, human CNS stem cells migrate away from the site of injection and differentiate into neurons and astrocytes. No tumor formation was ever observed. Aside from depending on scarce human neural fetal tissue, the use of human embryonic CNS stem cells for clinical neural transplantation should provide a reliable solution to some of the major problems that pertain to this field, and should allow determination of the safety characteristics of the donor cells in terms of tumorigenicity, viability, sterility, and antigenic compatibility far in advance of the scheduled day of surgery.


Asunto(s)
Diencéfalo/trasplante , Trasplante de Células Madre , Trasplante de Tejido Encefálico , Células Cultivadas , Sistema Nervioso Central/cirugía , Diencéfalo/citología , Trasplante de Tejido Fetal , Técnica del Anticuerpo Fluorescente , Humanos , Neuronas/citología , Células Madre/citología
13.
Neuroreport ; 10(2): 345-51, 1999 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-10203333

RESUMEN

The expression and regulation of matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs (TIMPs) in neuroectodermal precursor cells is undocumented. We report the presence of MMP-2, but no MMP-9, and of all the four known TIMPs in neuroepithelial stem cells isolated from the human CNS. The expression of TIMP-1, TIMP-2 and TIMP-3 was unchanged following stem cells differentiation into neurons and glia. In contrast, while MMP-2 and TIMP-4 were localized to both stem and mature CNS cells, their levels of expression were substantially reduced in the latter. TIMP-4 showed a 23-fold reduction in media conditioned by differentiated cells compared with stem cell-conditioned media, reflecting a 6-fold decrease in mRNA expression. Interestingly, TIMP-4 also differed from the other TIMPs in that it was cell-associated in the stem cells, where this fraction remained unchanged upon differentiation. Hence, regulation of selective MMPs and TIMPs occurs during differentiation of human neural precursors suggesting that MMP-2 and TIMP-4 in particular may perform regulatory roles in the developing CNS.


Asunto(s)
Diencéfalo/embriología , Gelatinasas/metabolismo , Metaloendopeptidasas/metabolismo , Células Madre/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Células Cultivadas , Colagenasas/genética , Colagenasas/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Gelatinasas/genética , Humanos , Metaloproteinasa 2 de la Matriz , Metaloproteinasa 9 de la Matriz , Metaloendopeptidasas/genética , ARN Mensajero/metabolismo , Inhibidores Tisulares de Metaloproteinasas/genética
14.
Neuroreport ; 5(18): 2489-92, 1994 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-7696587

RESUMEN

The presence of functional receptors for calcitonin gene-related peptide (CGRP) in the brain of adult rats and on nerve cell cultures was investigated. Neuronal and glial cultures were obtained from mesencephalons of embryos at gestational day 16. The response to CGRP was tested by measuring the adenylyl cyclase (AC) activity on isolated membranes. CGRP binding in adult rat brains was ineffective in activating AC, whereas a dose-dependent stimulation of AC activity was induced by the peptide both in neuronal and glial cultures. This effect was more pronounced in the glial cells where high affinity binding sites for CGRP were detected. The presence of functional CGRP receptors in embryonic mesencephalic cells, suggests a role for CGRP in the development of rat mesencephalon.


Asunto(s)
Adenilil Ciclasas/metabolismo , Encéfalo/embriología , Encéfalo/enzimología , Péptido Relacionado con Gen de Calcitonina/farmacología , Animales , Sitios de Unión , Encéfalo/efectos de los fármacos , Células Cultivadas , Activación Enzimática , Masculino , Ratas , Ratas Sprague-Dawley , Distribución Tisular
15.
Neurosci Lett ; 185(3): 151-4, 1995 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-7753479

RESUMEN

Stem cells isolated from the CNS of both embryonic and adult mice undergo extensive proliferation in the presence of epidermal growth factor (EGF). Removal of EGF determines the differentiation of these cells into neurons and glia. We have recently demonstrated that basic fibroblast growth factor (bFGF) regulates the proliferation of EGF-generated progenitors of the embryonic mouse striatum. We report here that bFGF induces proliferation of some EGF-generated precursors of the adult mouse striatum which, in turn, differentiate in vitro into cells possessing neuron-like morphology and neuronal antigenic properties. These results demonstrate that EGF and bFGF can act sequentially to regulate the de novo generation of neurons from the adult mouse CNS in vitro and suggest the existence of a lineage relationship between EGF- and bFGF-responsive progenitor cells of the adult murine brain.


Asunto(s)
Factor de Crecimiento Epidérmico/fisiología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células Madre/fisiología , Animales , División Celular , Sistema Nervioso Central/fisiología , Cuerpo Estriado , Ratones
16.
Biomed Pharmacother ; 55(4): 201-5, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11393806

RESUMEN

Stem cells play a critical role during embryo and tissue formation throughout development. Thanks to their multipotentiality - i.e., the ability to give rise to different lineages of mature cells - and to their extensive capacity for self-renewal and expansive growth, stem cells can also contribute to the maintenance of tissue integrity in adulthood. Historically, it has been held that fetal and adult (somatic) stem cells are tissue-specific 'entities' whose differentiation potential is limited to the generation of mature cell types of the tissue/organ in which they reside. Yet, recent years have seen the publication of an impressive sequence of reports dealing with what is now emerging as one of the most striking functional attributes of somatic stem cells, that is, their capacity to undergo transdifferentiation. Thanks to this peculiar characteristic adult stem cells display an unexpected ability to give rise to differentiated cells of tissues and organs different from those in which they reside. This commentary briefly illustrates the characteristics of the neural stem cell and its capacity as a neuroectodermal derivative to undergo transdifferentiation, thus giving rise to differentiated cells that normally originate from the mesoderm, like blood or skeletal muscle cells.


Asunto(s)
Diferenciación Celular , Neuronas/fisiología , Células Madre/fisiología , Animales , Humanos , Plasticidad Neuronal
17.
Cell Death Dis ; 4: e906, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24201805

RESUMEN

Mucopolysaccharidosis type II (MPSII or Hunter Syndrome) is a lysosomal storage disorder caused by the deficit of iduronate 2-sulfatase (IDS) activity and characterized by progressive systemic and neurological impairment. As the early mechanisms leading to neuronal degeneration remain elusive, we chose to examine the properties of neural stem cells (NSCs) isolated from an animal model of the disease in order to evaluate whether their neurogenic potential could be used to recapitulate the early phases of neurogenesis in the brain of Hunter disease patients. Experiments here reported show that NSCs derived from the subventricular zone (SVZ) of early symptomatic IDS-knockout (IDS-ko) mouse retained self-renewal capacity in vitro, but differentiated earlier than wild-type (wt) cells, displaying an evident lysosomal aggregation in oligodendroglial and astroglial cells. Consistently, the SVZ of IDS-ko mice appeared similar to the wt SVZ, whereas the cortex and striatum presented a disorganized neuronal pattern together with a significant increase of glial apoptotic cells, suggesting that glial degeneration likely precedes neuronal demise. Interestingly, a very similar pattern was observed in the brain cortex of a Hunter patient. These observations both in vitro, in our model, and in vivo suggest that IDS deficit seems to affect the late phases of neurogenesis and/or the survival of mature cells rather than NSC self-renewal. In particular, platelet-derived growth factor receptor-α-positive (PDGFR-α+) glial progenitors appeared reduced in both the IDS-ko NSCs and in the IDS-ko mouse and human Hunter brains, compared with the respective healthy controls. Treatment of mutant NSCs with IDS or PDGF throughout differentiation was able to increase the number of PDGFR-α+ cells and to reduce that of apoptotic cells to levels comparable to wt. This evidence supports IDS-ko NSCs as a reliable in vitro model of the disease, and suggests the rescue of PDGFR-α+ glial cells as a therapeutic strategy to prevent neuronal degeneration.


Asunto(s)
Mucopolisacaridosis II/metabolismo , Mucopolisacaridosis II/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Enfermedades Neurodegenerativas/patología , Neuroglía/metabolismo , Neuroglía/patología , Animales , Apoptosis/genética , Apoptosis/fisiología , Encéfalo/metabolismo , Encéfalo/patología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Glicoproteínas/deficiencia , Glicoproteínas/genética , Glicoproteínas/metabolismo , Enfermedades por Almacenamiento Lisosomal/metabolismo , Enfermedades por Almacenamiento Lisosomal/patología , Ratones , Ratones Noqueados , Mucopolisacaridosis II/genética , Enfermedades Neurodegenerativas/metabolismo
19.
Oncogene ; 28(15): 1807-11, 2009 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-19287454

RESUMEN

Glioblastomas (GBMs) contain transformed, self-maintaining, multipotent, tumour-initiating cancer stem cells, whose identification has radically changed our perspective on the physiology of these tumours. Currently, it is unknown whether multiple types of transformed precursors, which display alternative sets of the complement of properties of true cancer stem cells, can be found in a GBM. If different subsets of such cancer stem-like cells (CSCs) do exist, they might represent distinct cell targets, with a differential therapeutic importance, also depending on their characteristics and lineage relationship. Here, we report the presence of two types of CSCs within different regions of the same human GBM. Cytogenetic and molecular analysis shows that the two types of CSCs bear quite diverse tumorigenic potential and distinct genetic anomalies, and, yet, derive from common ancestor cells. This provides critical information to unravel the development of CSCs and the key molecular/genetic components underpinning tumorigenicity in human GBMs.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Glioblastoma/genética , Glioblastoma/patología , Células Madre Neoplásicas/patología , Animales , Línea Celular Tumoral , Aberraciones Cromosómicas , Genoma , Humanos , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad
20.
Artículo en Inglés | MEDLINE | ID: mdl-17939295

RESUMEN

Human glioblastomas appear to be established and expanded by cancer stem cells, which are endowed with tumour-initiating and perpetuating ability. We report that bone morphogenetic proteins (BMPs), amongst which BMP4 elicits the strongest effect, activate their cognate receptors (BMPRs) and trigger the Smad but not the MAP38 kinase signalling cascade in cells isolated from human glioblastomas (GBMs). This is followed by a reduction in proliferation and increased expression of differentiated neural markers, without affecting cell viability. The concomitant reduction in the clonogenic ability, both in the size of the CD133+ side population and in the growth kinetics of GBM cells, indicates that BMP4 triggers a reduction in the in vitro cancer stem cell (CSC) pool. Accordingly, transient ex vivo exposure to BMP4 abolishes the capacity of transplanted GBM cells to establish intracerebral GBMs. Most important, in vivo delivery of BMP4 effectively blocks the tumour growth and associated mortality which occur in 100% of control mice in less than 12 weeks, following intracerebral grafting of human GBM cells. These findings show that the BMP-BMPR signalling system, which controls the activity of normal brain stem cells, may also act as a key inhibitory regulator of cancer-initiating, GBM stem-like cells and identifies BMP4 as a novel, non-cytotoxic therapeutic effector, which may be used to prevent growth and recurrence of GBMs in humans.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Neoplasias Encefálicas/patología , Transformación Celular Neoplásica , Glioblastoma/patología , Células Madre/citología , Animales , Citometría de Flujo , Humanos , Inmunohistoquímica , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA