Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
BMC Neurosci ; 15: 100, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25156632

RESUMEN

BACKGROUND: Accumulating evidence suggests that growth hormone (GH) may play a major role in the regulation of postnatal neurogenesis, thus supporting the possibility that it may be also involved in promoting brain repair after brain injury. In order to gain further insight on this possibility, in this study we have investigated the pathways signaling the effect of GH treatment on the proliferation and survival of hippocampal subgranular zone (SGZ)-derived neurospheres. RESULTS: Our results demonstrate that GH treatment promotes both proliferation and survival of SGZ neurospheres. By using specific chemical inhibitors we have been also able to demonstrate that GH treatment promotes the activation of both Akt-mTOR and JNK signaling pathways, while blockade of these pathways either reduces or abolishes the GH effects. In contrast, no effect of GH on the activation of the Ras-ERK pathway was observed after GH treatment, despite blockade of this signaling path also resulted in a significant reduction of GH effects. Interestingly, SGZ cells were also capable of producing GH, and blockade of endogenous GH also resulted in a decrease in the proliferation and survival of SGZ neurospheres. CONCLUSIONS: Altogether, our findings suggest that GH treatment may promote the proliferation and survival of neural progenitors. This effect may be elicited by cooperating with locally-produced GH in order to increase the response of neural progenitors to adequate stimuli. On this view, the possibility of using GH treatment to promote neurogenesis and cell survival in some acquired neural injuries may be envisaged.


Asunto(s)
Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Hormona del Crecimiento/metabolismo , Hipocampo/fisiología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hormona del Crecimiento/antagonistas & inhibidores , Hipocampo/efectos de los fármacos , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones Endogámicos C57BL , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Proteínas ras/antagonistas & inhibidores , Proteínas ras/metabolismo
2.
Stem Cells ; 30(4): 773-84, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22893458

RESUMEN

Neural stem/progenitor cells present in the subventricular zone (SVZ) are a potential source of repairing cells after injury. Therefore, the identification of novel players that modulate neural stem cells differentiation can have a huge impact in stem cell-based therapies. Herein, we describe a unique role of histamine in inducing functional neuronal differentiation from cultured mouse SVZ stem/progenitor cells. This proneurogenic effect depends on histamine 1 receptor activation and involves epigenetic modifications and increased expression of Mash1, Dlx2, and Ngn1 genes. Biocompatible poly (lactic-co-glycolic acid) microparticles, engineered to release histamine in a controlled and prolonged manner, also triggered robust neuronal differentiation in vitro. Preconditioning with histamine-loaded microparticles facilitated neuronal differentiation of SVZ-GFP cells grafted in hippocampal slices and in in vivo rodent brain. We propose that neuronal commitment triggered by histamine per se or released from biomaterial-derived vehicles may represent a new tool for brain repair strategies.


Asunto(s)
Histamina/farmacología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Histamina/administración & dosificación , Histamina/química , Ácido Láctico/administración & dosificación , Ácido Láctico/química , Ventrículos Laterales/citología , Ventrículos Laterales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Ácido Poliglicólico/administración & dosificación , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
3.
J Neurochem ; 120(1): 93-105, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22007767

RESUMEN

Increasing evidences suggest that neuropeptide Y (NPY) may act as a key modulator of the cross-talk between the brain and the immune system in health and disease. In the present study, we dissected the possible inhibitory role of NPY upon inflammation-associated microglial cell motility. NPY, through activation of Y(1) receptors, was found to inhibit lipopolysaccharide (LPS)-induced microglia (N9 cell line) motility. Moreover, stimulation of microglia with LPS was inhibited by IL-1 receptor antagonist (IL-1ra), suggesting the involvement of endogenous interleukin-1 beta (IL-1ß) in this process. Direct stimulation with IL-1ß promoted downstream p38 mitogen-activated protein kinase mobilization and increased microglia motility. Moreover, consistently, p38 mitogen-activated protein kinase inhibition decreased the extent of actin filament reorganization occurring during plasma membrane ruffling and p38 phosphorylation was inhibited by NPY, involving Y(1) receptors. Significantly, the key inhibitory role of NPY on LPS-induced motility of CD11b-positive cells was further confirmed in mouse brain cortex explants. In summary, we revealed a novel functional role for NPY in the regulation of microglial function that may have important implications in the modulation of CNS injuries/diseases where microglia migration/motility might play a role.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/farmacología , Microglía/efectos de los fármacos , Neuropéptido Y/farmacología , Animales , Western Blotting , Antígeno CD11b/metabolismo , Línea Celular , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/ultraestructura , Interpretación Estadística de Datos , Inmunohistoquímica , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Receptores de Neuropéptido Y/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
4.
Eur J Neurosci ; 35(11): 1672-83, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22506985

RESUMEN

Ampakines are chemical compounds known to modulate the properties of ionotropic α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA)-subtype glutamate receptors. The functional effects attributed to ampakines involve plasticity and the increase in synaptic efficiency of neuronal circuits, a process that may be intimately associated with differentiation of newborn neurons. The subventricular zone (SVZ) is the main neurogenic niche of the brain, containing neural stem cells with brain repair potential. Accordingly, the identification of new pharmaceutical compounds with neurogenesis-enhancing properties is important as a tool to promote neuronal replacement based on the use of SVZ cells. The purpose of the present paper is to examine the possible proneurogenic effects of ampakine CX546 in cell cultures derived from the SVZ of early postnatal mice. We observed that CX546 (50 µm) treatment triggered an increase in proliferation, evaluated by BrdU incorporation assay, in the neuroblast lineage. Moreover, by using a cell viability assay (TUNEL) we found that, in contrast to AMPA, CX546 did not cause cell death. Also, both AMPA and CX546 stimulated neuronal differentiation as evaluated morphologically through neuronal nuclear protein (NeuN) immunocytochemistry and functionally by single-cell calcium imaging. Accordingly, short exposure to CX546 increased axonogenesis, as determined by the number and length of tau-positive axons co-labelled for the phosphorylated form of SAPK/JNK (P-JNK), and dendritogenesis (MAP2-positive neurites). Altogether, this study shows that ampakine CX546 promotes neurogenesis in SVZ cell cultures and thereby may have potential for future stem cell-based therapies.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Dioxoles/farmacología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Piperidinas/farmacología , Telencéfalo/citología , Telencéfalo/embriología , Animales , Animales Recién Nacidos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Cultivo Primario de Células
5.
J Neuroinflammation ; 9: 90, 2012 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-22569158

RESUMEN

BACKGROUND: Histamine is commonly acknowledged as an inflammatory mediator in peripheral tissues, leaving its role in brain immune responses scarcely studied. Therefore, our aim was to uncover the cellular and molecular mechanisms elicited by this molecule and its receptors in microglia-induced inflammation by evaluating cell migration and inflammatory mediator release. METHODS: Firstly, we detected the expression of all known histamine receptor subtypes (H1R, H2R, H3R and H4R), using a murine microglial cell line and primary microglia cell cultures from rat cortex, by real-time PCR analysis, immunocytochemistry and Western blotting. Then, we evaluated the role of histamine in microglial cell motility by performing scratch wound assays. Results were further confirmed using murine cortex explants. Finally, interleukin-1beta (IL-1ß) and tumor necrosis factor-alpha (TNF-α) levels were evaluated by ELISA measurements to determine the role of histamine on the release of these inflammatory mediators. RESULTS: After 12 h of treatment, 100 µM histamine and 10 µg/ml histamine-loaded poly (lactic-co-glycolic acid) microparticles significantly stimulated microglia motility via H4R activation. In addition, migration involves α5ß1 integrins, and p38 and Akt signaling pathways. Migration of microglial cells was also enhanced in the presence of lipopolysaccharide (LPS, 100 ng/ml), used as a positive control. Importantly, histamine inhibited LPS-stimulated migration via H4R activation. Histamine or H4R agonist also inhibited LPS-induced IL-1ß release in both N9 microglia cell line and hippocampal organotypic slice cultures. CONCLUSIONS: To our knowledge, we are the first to show a dual role of histamine in the modulation of microglial inflammatory responses. Altogether, our data suggest that histamine per se triggers microglia motility, whereas histamine impedes LPS-induced microglia migration and IL-1ß release. This last datum assigns a new putative anti-inflammatory role for histamine, acting via H4R to restrain exacerbated microglial responses under inflammatory challenge, which could have strong repercussions in the treatment of CNS disorders accompanied by microglia-derived inflammation.


Asunto(s)
Histamina/fisiología , Mediadores de Inflamación/fisiología , Microglía/fisiología , Receptores Acoplados a Proteínas G/fisiología , Receptores Histamínicos/fisiología , Animales , Línea Celular , Movimiento Celular/fisiología , Células Cultivadas , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiología , Hipocampo/metabolismo , Hipocampo/patología , Histamina/farmacología , Mediadores de Inflamación/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/metabolismo , Interleucina-1beta/fisiología , Lipopolisacáridos/farmacología , Lipopolisacáridos/fisiología , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Técnicas de Cultivo de Órganos , Ratas , Receptores Acoplados a Proteínas G/agonistas , Receptores Histamínicos H4
6.
J Neuroinflammation ; 9: 204, 2012 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-22901528

RESUMEN

BACKGROUND AND PURPOSE: Blockade of adenosine A(2A) receptors (A(2A)R) affords robust neuroprotection in a number of brain conditions, although the mechanisms are still unknown. A likely candidate mechanism for this neuroprotection is the control of neuroinflammation, which contributes to the amplification of neurodegeneration, mainly through the abnormal release of pro-inflammatory cytokines such as interleukin(IL)-1ß. We investigated whether A(2A)R controls the signaling of IL-1ß and its deleterious effects in cultured hippocampal neurons. METHODS: Hippocampal neuronal cultures were treated with IL-1ß and/or glutamate in the presence or absence of the selective A(2A)R antagonist, SCH58261 (50 nmol/l). The effect of SCH58261 on the IL-1ß-induced phosphorylation of the mitogen-activated protein kinases (MAPKs) c-Jun N-terminal kinase (JNK) and p38 was evaluated by western blotting and immunocytochemistry. The effect of SCH58261 on glutamate-induced neurodegeneration in the presence or absence of IL-1ß was evaluated by nucleic acid and by propidium iodide staining, and by lactate dehydrogenase assay. Finally, the effect of A(2A)R blockade on glutamate-induced intracellular calcium, in the presence or absence of IL-1ß, was studied using single-cell calcium imaging. RESULTS: IL-1ß (10 to 100 ng/ml) enhanced both JNK and p38 phosphorylation, and these effects were prevented by the IL-1 type 1 receptor antagonist IL-1Ra (5 µg/ml), in accordance with the neuronal localization of IL-1 type 1 receptors, including pre-synaptically and post-synaptically. At 100 ng/ml, IL-1ß failed to affect neuronal viability but exacerbated the neurotoxicity induced by treatment with 100 µmol/l glutamate for 25 minutes (evaluated after 24 hours). It is likely that this resulted from the ability of IL-1ß to enhance glutamate-induced calcium entry and late calcium deregulation, both of which were unaffected by IL-1ß alone. The selective A(2A)R antagonist, SCH58261 (50 nmol/l), prevented both the IL-1ß-induced phosphorylation of JNK and p38, as well as the IL-1ß-induced deregulation of calcium and the consequent enhanced neurotoxicity, whereas it had no effect on glutamate actions. CONCLUSIONS: These results prompt the hypothesis that the neuroprotection afforded by A(2A)R blockade might result from this particular ability of A(2A)R to control IL-1ß-induced exacerbation of excitotoxic neuronal damage, through the control of MAPK activation and late calcium deregulation.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Interleucina-1beta/toxicidad , Sistema de Señalización de MAP Quinasas/fisiología , Neuronas/metabolismo , Receptor de Adenosina A2A/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Células Cultivadas , Femenino , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Neuronas/efectos de los fármacos , Neuronas/patología , Embarazo , Pirimidinas/farmacología , Ratas , Ratas Wistar , Triazoles/farmacología
7.
J Neurosci ; 30(13): 4573-84, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20357108

RESUMEN

In the adult mammalian brain, the subventricular zone (SVZ) hosts stem cells constantly generating new neurons. Angiopoietin-1 (Ang-1) is an endothelial growth factor with a critical role in division, survival, and adhesion of endothelial cells via Tie-2 receptor activity. Expression of Tie-2 in nonendothelial cells, especially neurons and stem cells, suggests that Ang-1 may be involved in neurogenesis. In the present work, we investigated the putative role of Ang-1 on SVZ neurogenesis. Immature cells from SVZ-derived neurospheres express Ang-1 and Tie-2 mRNA, suggesting a role for the Ang-1/Tie-2 system in the neurogenic niche. Moreover, we also found that Tie-2 protein expression is retained on differentiation in neurons and glial cells. Ang-1 triggered proliferation via activation of the ERK1/2 (extracellular signal-regulated kinase 1/2) mitogen-activated protein kinase (MAPK) kinase pathway but did not induce cell death. Accordingly, coincubation with an anti-Tie-2 neutralizing antibody prevented the pro-proliferative effect of Ang-1. Furthermore, Ang-1 increased the number of NeuN (neuronal nuclear protein)-positive neurons in cultures treated for 7 d, as well as the number of functional neurons, as assessed by monitoring [Ca(2+)](i) rises after application of specific stimuli for neurons and immature cells. The proneurogenic effect of Ang-1 is mediated by Tie-2 activation and subsequent mTOR (mammalian target of rapamycin kinase) mobilization. In agreement, neuronal differentiation significantly decreased after exposure to an anti-Tie-2 neutralizing antibody and to rapamycin. Moreover, Ang-1 elicited the activation of the SAPK (stress-activated protein kinase)/JNK (c-Jun N-terminal kinase) MAPK, involved in axonogenesis. Our work shows a proneurogenic effect of Ang-1, highlighting the relevance of blood vessel/stem cell cross talk in health and disease.


Asunto(s)
Angiopoyetina 1/fisiología , Encéfalo/citología , Neuronas/fisiología , Células Madre/fisiología , Angiopoyetina 1/biosíntesis , Angiopoyetina 1/genética , Animales , Axones/fisiología , Encéfalo/crecimiento & desarrollo , Muerte Celular , Diferenciación Celular , Proliferación Celular , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/crecimiento & desarrollo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Neurogénesis , Neuroglía/citología , Neuroglía/fisiología , Neuronas/citología , Bulbo Olfatorio/citología , Bulbo Olfatorio/crecimiento & desarrollo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/biosíntesis , Receptor TIE-2/biosíntesis , Receptor TIE-2/genética , Células Madre/citología , Serina-Treonina Quinasas TOR
8.
J Neurochem ; 116(6): 1018-27, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21175616

RESUMEN

The subventricular zone (SVZ) is a major reservoir for stem cells in the adult mammalian brain. Neural stem cells supply the olfactory bulb with new interneurons and provide cells that migrate towards lesioned brain areas. Neuropeptide Y (NPY), one of the most abundant neuropeptides in the brain, was previously shown to induce neuroproliferation on mice SVZ cells. In the present study, performed in rats, we demonstrate the endogenous synthesis of NPY by cells in the SVZ that suggests that NPY could act as an autocrine/paracrine factor within the SVZ area. We observed that NPY promotes SVZ cell proliferation as previously reported in mice, but does not affect self-renewal of SVZ stem cells. Additionally, this study provides the first direct evidence of a chemokinetic activity of NPY on SVZ cells. Using pharmacological approaches, we demonstrate that both the mitogenic and chemokinetic properties of NPY involve Y1 receptor-mediated activation of the ERK1/2 MAP kinase pathway. Altogether, our data establish that NPY through Y1 receptors activation controls chemokinetic activity and, as for mice, is a major neuroproliferative regulator of rat SVZ cells.


Asunto(s)
Movimiento Celular/fisiología , Ventrículos Cerebrales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuronas/fisiología , Neuropéptido Y/metabolismo , Animales , Animales Recién Nacidos , Arginina/análogos & derivados , Arginina/farmacología , Bromodesoxiuridina/metabolismo , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuropéptido Y/análogos & derivados , Neuropéptido Y/genética , Neuropéptido Y/farmacología , Fragmentos de Péptidos/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/genética , Receptores de Neuropéptido/metabolismo
9.
Stem Cells ; 28(7): 1219-30, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20506358

RESUMEN

Nitric oxide (NO) was described to inhibit the proliferation of neural stem cells. Some evidence suggests that NO, under certain conditions, can also promote cell proliferation, although the mechanisms responsible for a potential proliferative effect of NO in neural stem cells have remained unaddressed. In this work, we investigated and characterized the proliferative effect of NO in cell cultures obtained from the mouse subventricular zone. We found that the NO donor NOC-18 (10 microM) increased cell proliferation, whereas higher concentrations (100 microM) inhibited cell proliferation. Increased cell proliferation was detected rapidly following exposure to NO and was prevented by blocking the mitogen-activated kinase (MAPK) pathway, independently of the epidermal growth factor (EGF) receptor. Downstream of the EGF receptor, NO activated p21Ras and the MAPK pathway, resulting in a decrease in the nuclear presence of the cyclin-dependent kinase inhibitor 1, p27(KIP1), allowing for cell cycle progression. Furthermore, in a mouse model that shows increased proliferation of neural stem cells in the hippocampus following seizure injury, we observed that the absence of inducible nitric oxide synthase (iNOS(-/-) mice) prevented the increase in cell proliferation observed following seizures in wild-type mice, showing that NO from iNOS origin is important for increased cell proliferation following a brain insult. Overall, we show that NO is able to stimulate the proliferation of neural stem cells bypassing the EGF receptor and promoting cell division. Moreover, under pathophysiological conditions in vivo, NO from iNOS origin also promotes proliferation in the hippocampus.


Asunto(s)
Proliferación Celular , Receptores ErbB/metabolismo , Neuronas/citología , Neuronas/metabolismo , Óxido Nítrico/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Células Cultivadas , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/metabolismo
10.
STAR Protoc ; 2(1): 100382, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33748784

RESUMEN

BDNF levels are reduced in the chronically stressed brain, in the area of hippocampus. Part of the hippocampal BDNF is provided by neuronal projection of the entorhinal cortex. Studying the cortico-hippocampal transport of BDNF in vivo is technically difficult. Here, we describe a protocol that reproduces mouse cortico-hippocampal circuit in vitro by plating neurons on the microfluidic devices and infecting the neurons with virus-encoding BDNF-mCherry, which allows investigation of the effects of elevated corticosterone levels on BDNF axonal transport. For complete details on the use and execution of this protocol, please refer to Agasse et al. (2020).


Asunto(s)
Transporte Axonal/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Animales , Axones/fisiología , Encéfalo/fisiología , Corticosterona/farmacología , Corteza Entorrinal/fisiología , Glucocorticoides/farmacología , Hipocampo/fisiología , Dispositivos Laboratorio en un Chip , Ratones , Microfluídica/métodos , Red Nerviosa/fisiología , Neuronas/metabolismo , Transporte de Proteínas/fisiología
11.
Stem Cells ; 27(2): 408-19, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18988709

RESUMEN

Neural stem cells persist in the adult mammalian brain, within the subventricular zone (SVZ). The endogenous mechanisms underpinning SVZ neural stem cell proliferation, self-renewal, and differentiation are not fully elucidated. In the present report, we describe a growth-stimulatory activity of liver explant-conditioned media on SVZ cell cultures and identify hepatocyte growth factor (HGF) as a major player in this effect. HGF exhibited a mitogenic activity on SVZ cell cultures in a mitogen-activated protein kinase (MAPK) (ERK1/2)-dependent manner as U0126, a specific MAPK inhibitor, blocked it. Combining a functional neurosphere forming assay with immunostaining for c-Met, along with markers of SVZ cells subtypes, demonstrated that HGF promotes the expansion of neural stem-like cells that form neurospheres and self-renew. Immunostaining, HGF enzyme-linked immunosorbent assay and Madin-Darby canine kidney cell scattering assay indicated that SVZ cell cultures produce and release HGF. SVZ cell-conditioned media induced proliferation on SVZ cell cultures, which was blocked by HGF-neutralizing antibodies, hence implying that endogenously produced HGF accounts for a major part in SVZ mitogenic activity. Brain sections immunostaining revealed that HGF is produced by nestin-expressing cells and c-Met is expressed within the SVZ by immature cells. HGF intracerebroventricular injection promoted SVZ cell proliferation and increased the ability of these cells exposed in vivo to HGF to form neurospheres in vitro, whereas intracerebroventricular injection of HGF-neutralizing antibodies decreased SVZ cell proliferation. The present study unravels a major role, both in vitro and in vivo, for endogenous HGF in SVZ neural stem cell growth and self-renewal.


Asunto(s)
Ventrículos Cerebrales/citología , Factor de Crecimiento de Hepatocito/fisiología , Neuronas/citología , Transducción de Señal/fisiología , Células Madre/citología , Animales , Diferenciación Celular , Línea Celular , Proliferación Celular , Células Cultivadas , Perros , Ensayo de Inmunoadsorción Enzimática , Factor de Crecimiento de Hepatocito/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar
12.
Cell Rep ; 32(1): 107865, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32640230

RESUMEN

Chronic exposure to stress is a major risk factor for neuropsychiatric disease, and elevated plasma corticosterone (CORT) correlates with reduced levels of both brain-derived neurotrophic factor (BDNF) and hippocampal neurogenesis. Precisely how these phenomena are linked, however, remains unclear. Using a cortico-hippocampal network-on-a-chip, we find that the glucocorticoid receptor agonist dexamethasone (DXM) stimulates the cyclin-dependent kinase 5 (CDK5) to phosphorylate huntingtin (HTT) at serines 1181 and 1201 (S1181/1201), which retards BDNF vesicular transport in cortical axons. Parallel studies in mice show that CORT induces phosphorylation of these same residues, reduces BDNF levels, and suppresses neurogenesis. The adverse effects of CORT are reduced in mice bearing an unphosphorylatable mutant HTT (HdhS1181A/S1201A). The protective effect of unphosphorylatable HTT, however, disappears if neurogenesis is blocked. The CDK5-HTT pathway, which regulates BDNF transport in the cortico-hippocampal network, thus provides a missing link between elevated CORT levels and suppressed neurogenesis.


Asunto(s)
Envejecimiento/metabolismo , Corticosterona/metabolismo , Hipocampo/metabolismo , Proteína Huntingtina/metabolismo , Neurogénesis , Animales , Conducta Animal , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Depresión/metabolismo , Femenino , Masculino , Ratones Endogámicos C57BL , Fosforilación , Transporte de Proteínas
13.
Science ; 369(6505): 787-793, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32675289

RESUMEN

Although Huntington's disease is a late-manifesting neurodegenerative disorder, both mouse studies and neuroimaging studies of presymptomatic mutation carriers suggest that Huntington's disease might affect neurodevelopment. To determine whether this is actually the case, we examined tissue from human fetuses (13 weeks gestation) that carried the Huntington's disease mutation. These tissues showed clear abnormalities in the developing cortex, including mislocalization of mutant huntingtin and junctional complex proteins, defects in neuroprogenitor cell polarity and differentiation, abnormal ciliogenesis, and changes in mitosis and cell cycle progression. We observed the same phenomena in Huntington's disease mouse embryos, where we linked these abnormalities to defects in interkinetic nuclear migration of progenitor cells. Huntington's disease thus has a neurodevelopmental component and is not solely a degenerative disease.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Sistema Nervioso/embriología , Animales , Ciclo Celular , Endosomas/metabolismo , Feto , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Ratones , Ratones Mutantes , Mitosis , Mutación , Células Neuroepiteliales/metabolismo , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
14.
Stem Cells ; 26(9): 2361-71, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18583543

RESUMEN

Tumor necrosis factor (TNF)-alpha has been reported to modulate brain injury, but remarkably, little is known about its effects on neurogenesis. We report that TNF-alpha strongly influences survival, proliferation, and neuronal differentiation in cultured subventricular zone (SVZ) neural stem/progenitor cells derived from the neonatal P1-3 C57BL/6 mice. By using single-cell calcium imaging, we developed a method, based on cellular response to KCl and/or histamine, that allows the functional evaluation of neuronal differentiation. Exposure of SVZ cultures to 1 and 10 ng/ml mouse or 1 ng/ml human recombinant TNF-alpha resulted in increased differentiation of cells displaying a neuronal-like profile of [Ca2+](i) responses, compared with the predominant profile of immature cells observed in control, nontreated cultures. Moreover, by using neutralizing antibodies for each TNF-alpha receptor, we found that the proneurogenic effect of 1 ng/ml TNF-alpha is mediated via tumor necrosis factor receptor 1 activation. Accordingly, the percentage of neuronal nuclear protein-positive neurons was increased following exposure to mouse TNF-alpha. Interestingly, exposure of SVZ cultures to 1 ng/ml TNF-alpha induced cell proliferation, whereas 10 and 100 ng/ml TNF-alpha induced apoptotic cell death. Moreover, we found that exposure of SVZ cells to TNF-alpha for 15 minutes or 6 hours caused an increase in the phospho-stress-activated protein kinase/c-Jun N-terminal kinase immunoreactivity initially in the nucleus and then in growing axons, colocalizing with tau, consistent with axonogenesis. Taken together, these results show that TNF-alpha induces neurogenesis in neonatal SVZ cell cultures of mice. TNF-alpha, a proinflammatory cytokine and a proneurogenic factor, may play a central role in promoting neurogenesis and brain repair in response to brain injury and infection.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neuronas/citología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Animales Recién Nacidos , Encéfalo/citología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Endogámicos C57BL , Neurogénesis , Neuronas/efectos de los fármacos , Proteínas Recombinantes/farmacología , Células Madre/citología , Células Madre/efectos de los fármacos
15.
Stem Cells ; 26(6): 1636-45, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18388302

RESUMEN

Stem cells of the subventricular zone (SVZ) represent a reliable source of neurons for cell replacement. Neuropeptide Y (NPY) promotes neurogenesis in the hippocampal subgranular layer and the olfactory epithelium and may be useful for the stimulation of SVZ dynamic in brain repair purposes. We describe that NPY promotes SVZ neurogenesis. NPY (1 microM) treatments increased proliferation at 48 hours and neuronal differentiation at 7 days in SVZ cell cultures. NPY proneurogenic properties are mediated via the Y1 receptor. Accordingly, Y1 receptor is a major active NPY receptor in the mouse SVZ, as shown by functional autoradiography. Moreover, short exposure to NPY increased immunoreactivity for the phosphorylated form of extracellular signal-regulated kinase 1/2 in the nucleus, compatible with a trigger for proliferation, whereas 6 hours of treatment amplified the phosphorylated form of c-Jun-NH(2)-terminal kinase signal in growing axons, consistent with axonogenesis. NPY, as a promoter of SVZ neurogenesis, is a crucial factor for future development of cell-based brain therapy. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Ventrículos Cerebrales/citología , Neuronas/citología , Neuropéptido Y/farmacología , Animales , Calcio/fisiología , Muerte Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Ventrículos Cerebrales/efectos de los fármacos , Ventrículos Cerebrales/fisiología , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/fisiología
16.
Rejuvenation Res ; 11(1): 187-200, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18279032

RESUMEN

Subventricular zone (SVZ) cell cultures contain mixed populations of immature cells, neurons, astrocytes, and progenitors in different stages of development. In the present work, we examined whether cell types of the SVZ could be functionally discriminated on the basis of intracellular free calcium level ([Ca(2+)](i)) variations following KCl and histamine stimulation. For this purpose, [Ca(2+)](i) were measured in SVZ cell cultures from neonatal P1-3 C57Bl/6 donor mice, in single cells, after stimulation with 100 microM histamine or 50 mM KCl. MAP-2-positive neurons and doublecortin-positive neuroblasts were distinguished on the basis of their selective ratio of response to KCl and/or histamine stimulation. Moreover, we could distinguish immature cells on the basis of the selective response to histamine via the histamine 1 receptor activation. Exposure of SVZ cultures to the pro-neurogenic stem cell factor (SCF) induced an increase in the number of cells responding to KCl and a decrease in the number of cells responding to histamine, consistent with neuronal differentiation. The selective response to KCl/histamine in single cell calcium imaging analysis offers a rapid and efficient way for the functional discrimination of neuronal differentiation in SVZ cell cultures, opening new perspectives for the search of potential pro-neurogenic factors.


Asunto(s)
Ventrículos Cerebrales/efectos de los fármacos , Histamina/farmacología , Neuronas/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Animales Recién Nacidos , Astrocitos , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Ventrículos Cerebrales/metabolismo , Ventrículos Cerebrales/fisiología , Histamina/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/fisiología , Cloruro de Potasio/farmacología , Receptores Histamínicos/metabolismo , Células Madre/metabolismo , Células Madre/fisiología
17.
Front Cell Neurosci ; 10: 284, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28018177

RESUMEN

Neurogenesis in the subventricular zone (SVZ) is regulated by diffusible factors and cell-cell contacts. In vivo, SVZ stem cells are associated with the abluminal surface of blood vessels and such interactions are thought to regulate their neurogenic capacity. SVZ neural stem cells (NSCs) have been described to contact endothelial-derived laminin via α6ß1 integrin. To elucidate whether heterocellular contacts with brain endothelial cells (BEC) regulate SVZ cells neurogenic capacities, cocultures of SVZ neurospheres and primary BEC, both obtained from C57BL/6 mice, were performed. The involvement of laminin-integrin interactions in SVZ homeostasis was tested in three ways. Firstly, SVZ cells were analyzed following incubation of BEC with the protein synthesis inhibitor cycloheximide (CHX) prior to coculture, a treatment expected to decrease membrane proteins. Secondly, SVZ cells were cocultured with BEC in the presence of an anti-α6 integrin neutralizing antibody. Thirdly, BEC were cultured with ß1-/- SVZ cells. We showed that contact with BEC supports, at least in part, proliferation and stemness of SVZ cells, as evaluated by the number of BrdU positive (+) and Sox2+ cells in contact with BEC. These effects are dependent on BEC-derived laminin binding to α6ß1 integrin and are decreased in cocultures incubated with anti-α6 integrin neutralizing antibody and in cocultures with SVZ ß1-/- cells. Moreover, BEC-derived laminin sustains stemness in SVZ cell cultures via activation of the Notch and mTOR signaling pathways. Our results show that BEC/SVZ interactions involving α6ß1 integrin binding to laminin, contribute to SVZ cell proliferation and stemness.

18.
Cancer Lett ; 371(2): 240-50, 2016 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-26683773

RESUMEN

Glioblastoma multiforme (GBM), a highly malignant brain tumor, accounts for half of all gliomas. Despite surgery, radiation and chemotherapy, the median survival is between 12 and 15 months. The poor prognosis is due to tumor recurrence attributed to chemoresistant glioma cancer stem cells (GSCs). Here we examined the effects of a novel compound NEO212, which is composed of two covalently conjugated anti-cancer compounds - temozolomide (TMZ) and perillyl alcohol (POH), on GSCs expressing either the proneural or mesenchymal gene signatures. These GSCs were obtained from patient-derived tumor tissue. Our findings demonstrate that NEO212 is 10 fold more cytotoxic to GSCs than TMZ (standard-of-care). Furthermore, NEO212 is effective against both proneural and clinically aggressive mesenchymal GSC subtypes. The mechanism of NEO212 mediated-cytotoxicity is through double-strand DNA breaks and apoptosis. In vivo studies show that NEO212 significantly delays tumor growth of both proneural and mesenchymal tumor stem cell populations. Patient-derived GSCs and tumors derived from these cells are highly reflective of the heterogeneity in human GBM. The efficacy of NEO212 against both GSC subtypes indicates that NEO212 has great clinical potential to effectively target GBM.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Glioma/tratamiento farmacológico , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Roturas del ADN de Doble Cadena , Dacarbazina/farmacología , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/metabolismo , Glioma/patología , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Temozolomida , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Stem Cell Res ; 13(2): 329-41, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25201326

RESUMEN

Methamphetamine (METH) is a highly addictive psychostimulant drug of abuse that negatively interferes with neurogenesis. In fact, we have previously shown that METH triggers stem/progenitor cell death and decreases neuronal differentiation in the dentate gyrus (DG). Still, little is known regarding its effect on DG stem cell properties. Herein, we investigate the impact of METH on mice DG stem/progenitor cell self-renewal functions. METH (10nM) decreased DG stem cell self-renewal, while 1nM delayed cell cycle in the G0/G1-to-S phase transition and increased the number of quiescent cells (G0 phase), which correlated with a decrease in cyclin E, pEGFR and pERK1/2 protein levels. Importantly, both drug concentrations (1 or 10nM) did not induce cell death. In accordance with the impairment of self-renewal capacity, METH (10nM) decreased Sox2(+)/Sox2(+) while increased Sox2(-)/Sox2(-) pairs of daughter cells. This effect relied on N-methyl-d-aspartate (NMDA) signaling, which was prevented by the NMDA receptor antagonist, MK-801 (10µM). Moreover, METH (10nM) increased doublecortin (DCX) protein levels consistent with neuronal differentiation. In conclusion, METH alters DG stem cell properties by delaying cell cycle and decreasing self-renewal capacities, mechanisms that may contribute to DG neurogenesis impairment followed by cognitive deficits verified in METH consumers.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/toxicidad , Giro Dentado/efectos de los fármacos , Metanfetamina/toxicidad , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Animales Recién Nacidos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Células Cultivadas , Ciclina E/metabolismo , Giro Dentado/metabolismo , Giro Dentado/patología , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Receptores ErbB/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , N-Metilaspartato/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neuronas/metabolismo , Neuronas/patología , Neuropéptidos/metabolismo , Fosforilación , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
20.
Front Cell Neurosci ; 8: 59, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24578683

RESUMEN

Neural stem cells (NSCs) from the subventricular zone (SVZ) have been indicated as a source of new oligodendrocytes to use in regenerative medicine for myelin pathologies. Indeed, NSCs are multipotent cells that can self-renew and differentiate into all neural cell types of the central nervous system. In normal conditions, SVZ cells are poorly oligodendrogenic, nevertheless their oligodendrogenic potential is boosted following demyelination. Importantly, progressive restriction into the oligodendrocyte fate is specified by extrinsic and intrinsic factors, endocannabinoids being one of these factors. Although a role for endocannabinoids in oligodendrogenesis has already been foreseen, selective agonists and antagonists of cannabinoids receptors produce severe adverse side effects. Herein, we show that hemopressin (Hp), a modulator of CB1 receptors, increased oligodendroglial differentiation in SVZ neural stem/progenitor cell cultures derived from neonatal mice. The original results presented in this work suggest that Hp and derivates may be of potential interest for the development of future strategies to treat demyelinating diseases.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA