Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Stem Cells ; 39(10): 1362-1381, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34043863

RESUMEN

Adenosine A2A receptor (A2A R) activation modulates several brain processes, ranging from neuronal maturation to synaptic plasticity. Most of these actions occur through the modulation of the actions of the neurotrophin brain-derived neurotrophic factor (BDNF). In this work, we studied the role of A2A Rs in regulating postnatal and adult neurogenesis in the rat hippocampal dentate gyrus (DG). Here, we show that A2A R activation with CGS 21680 promoted neural stem cell self-renewal, protected committed neuronal cells from cell death and contributed to a higher density of immature and mature neuronal cells, particularly glutamatergic neurons. Moreover, A2A R endogenous activation was found to be essential for BDNF-mediated increase in cell proliferation and neuronal differentiation. Our findings contribute to further understand the role of adenosinergic signaling in the brain and may have an impact in the development of strategies for brain repair under pathological conditions.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Hipocampo , Neurogénesis , Receptor de Adenosina A2A , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Ratas , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2A/metabolismo
2.
Stem Cells ; 39(9): 1253-1269, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33963799

RESUMEN

Although previous studies suggest that neural stem cells (NSCs) exist in the adult olfactory bulb (OB), their location, identity, and capacity to generate mature neurons in vivo has been little explored. Here, we injected enhanced green fluorescent protein (EGFP)-expressing retroviral particles into the OB core of adult mice to label dividing cells and to track the differentiation/maturation of any neurons they might generate. EGFP-labeled cells initially expressed adult NSC markers on days 1 to 3 postinjection (dpi), including Nestin, GLAST, Sox2, Prominin-1, and GFAP. EGFP+ -doublecortin (DCX) cells with a migratory morphology were also detected and their abundance increased over a 7-day period. Furthermore, EGFP-labeled cells progressively became NeuN+ neurons, they acquired neuronal morphologies, and they became immunoreactive for OB neuron subtype markers, the most abundant representing calretinin expressing interneurons. OB-NSCs also generated glial cells, suggesting they could be multipotent in vivo. Significantly, the newly generated neurons established and received synaptic contacts, and they expressed presynaptic proteins and the transcription factor pCREB. By contrast, when the retroviral particles were injected into the subventricular zone (SVZ), nearly all (98%) EGFP+ -cells were postmitotic when they reached the OB core, implying that the vast majority of proliferating cells present in the OB are not derived from the SVZ. Furthermore, we detected slowly dividing label-retaining cells in this region that could correspond to the population of resident NSCs. This is the first time NSCs located in the adult OB core have been shown to generate neurons that incorporate into OB circuits in vivo.


Asunto(s)
Células-Madre Neurales , Bulbo Olfatorio , Animales , Diferenciación Celular/fisiología , Interneuronas/metabolismo , Ratones , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/metabolismo
3.
J Neuroinflammation ; 18(1): 75, 2021 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-33736657

RESUMEN

BACKGROUND: Aging and age-related diseases are strong risk factors for the development of neurodegenerative diseases. Neuroinflammation (NIF), as the brain's immune response, plays an important role in aged associated degeneration of central nervous system (CNS). There is a need for well characterized animal models that will allow the scientific community to understand and modulate this process. METHODS: We have analyzed aging-phenotypical and inflammatory changes of brain myeloid cells (bMyC) in a senescent accelerated prone aged (SAMP8) mouse model, and compared with their senescence resistant control mice (SAMR1). We have performed morphometric methods to evaluate the architecture of cellular prolongations and determined the appearance of Iba1+ clustered cells with aging. To analyze specific constant brain areas, we have performed stereology measurements of Iba1+ cells in the hippocampal formation. We have isolated bMyC from brain parenchyma (BP) and choroid plexus plus meningeal membranes (m/Ch), and analyzed their response to systemic lipopolysaccharide (LPS)-driven inflammation. RESULTS: Aged 10 months old SAMP8 mice present many of the hallmarks of aging-dependent neuroinflammation when compared with their SAMR1 control, i.e., increase of protein aggregates, presence of Iba1+ clusters, but not an increase in the number of Iba1+ cells. We have further observed an increase of main inflammatory mediator IL-1ß, and an augment of border MHCII+Iba1+ cells. Isolated CD45+ bMyC from brain parenchyma (BP) and choroid plexus plus meningeal membranes (m/Ch) have been analyzed, showing that there is not a significant increase of CD45+ cells from the periphery. Our data support that aged-driven pro-inflammatory cytokine interleukin 1 beta (IL-1ß) transcription is enhanced in CD45+BP cells. Furthermore, LPS-driven systemic inflammation produces inflammatory cytokines mainly in border bMyC, sensed to a lesser extent by the BP bMyC, showing that IL-1ß expression is further augmented in aged SAMP8 compared to control SAMR1. CONCLUSION: Our data validate the SAMP8 model to study age-associated neuroinflammatory events, but careful controls for age and strain are required. These animals show morphological changes in their bMyC cell repertoires associated to age, corresponding to an increase in the production of pro-inflammatory cytokines such as IL-1ß, which predispose the brain to an enhanced inflammatory response after LPS-systemic challenge.


Asunto(s)
Envejecimiento Prematuro/genética , Envejecimiento/patología , Encefalitis/genética , Encefalitis/patología , Animales , Encéfalo/patología , Proteínas de Unión al Calcio/metabolismo , Plexo Coroideo/metabolismo , Plexo Coroideo/patología , Modelos Animales de Enfermedad , Encefalitis/inducido químicamente , Hipocampo/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos , Meninges/metabolismo , Meninges/patología , Ratones , Proteínas de Microfilamentos/metabolismo
4.
Proc Natl Acad Sci U S A ; 115(45): 11625-11630, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30352848

RESUMEN

Increasing age is the greatest known risk factor for the sporadic late-onset forms of neurodegenerative disorders such as Alzheimer's disease (AD). One of the brain regions most severely affected in AD is the hippocampus, a privileged structure that contains adult neural stem cells (NSCs) with neurogenic capacity. Hippocampal neurogenesis decreases during aging and the decrease is exacerbated in AD, but the mechanistic causes underlying this progressive decline remain largely unexplored. We here investigated the effect of age on NSCs and neurogenesis by analyzing the senescence accelerated mouse prone 8 (SAMP8) strain, a nontransgenic short-lived strain that spontaneously develops a pathological profile similar to that of AD and that has been employed as a model system to study the transition from healthy aging to neurodegeneration. We show that SAMP8 mice display an accelerated loss of the NSC pool that coincides with an aberrant rise in BMP6 protein, enhanced canonical BMP signaling, and increased astroglial differentiation. In vitro assays demonstrate that BMP6 severely impairs NSC expansion and promotes NSC differentiation into postmitotic astrocytes. Blocking the dysregulation of the BMP pathway and its progliogenic effect in vivo by intracranial delivery of the antagonist Noggin restores hippocampal NSC numbers, neurogenesis, and behavior in SAMP8 mice. Thus, manipulating the local microenvironment of the NSC pool counteracts hippocampal dysfunction in pathological aging. Our results shed light on interventions that may allow taking advantage of the brain's natural plastic capacity to enhance cognitive function in late adulthood and in chronic neurodegenerative diseases such as AD.


Asunto(s)
Envejecimiento/genética , Enfermedad de Alzheimer/tratamiento farmacológico , Proteína Morfogenética Ósea 6/genética , Proteínas Portadoras/farmacología , Células-Madre Neurales/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Células Madre Adultas/efectos de los fármacos , Células Madre Adultas/metabolismo , Células Madre Adultas/patología , Envejecimiento/metabolismo , Envejecimiento/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Proteína Morfogenética Ósea 6/antagonistas & inhibidores , Proteína Morfogenética Ósea 6/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Transducción de Señal
5.
Stem Cells ; 33(1): 219-29, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25185890

RESUMEN

Members of the cyclin-dependent kinase (CDK)-inhibitory protein (CIP)/kinase-inhibitory protein (KIP) family of cyclin-dependent kinase inhibitors regulate proliferation and cell cycle exit of mammalian cells. In the adult brain, the CIP/KIP protein p27(kip1) has been related to the regulation of intermediate progenitor cells located in neurogenic niches. Here, we uncover a novel function of p27(kip1) in the adult hippocampus as a dual regulator of stem cell quiescence and of cell-cycle exit of immature neurons. In vivo, p27(kip1) is detected in radial stem cells expressing SOX2 and in newborn neurons of the dentate gyrus. In vitro, the Cdkn1b gene encoding p27(kip1) is transcriptionally upregulated by quiescence signals such as BMP4. The nuclear accumulation of p27(kip1) protein in adult hippocampal stem cells encompasses the BMP4-induced quiescent state and its overexpression is able to block proliferation. p27(kip1) is also expressed in immature neurons upon differentiation of adult hippocampal stem cell cultures. Loss of p27(kip1) leads to an increase in proliferation and neurogenesis in the adult dentate gyrus, which results from both a decrease in the percentage of radial stem cells that are quiescent and a delay in cell cycle exit of immature neurons. Analysis of animals carrying a disruption in the cyclin-CDK interaction domain of p27(kip1) indicates that the CDK inhibitory function of the protein is necessary to control the activity of radial stem cells. Thus, we report that p27(kip1) acts as a central player of the molecular program that keeps adult hippocampal stem cells out of the cell cycle.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Hipocampo/citología , Células-Madre Neurales/citología , Neurogénesis/fisiología , Animales , Apoptosis/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Hipocampo/metabolismo , Humanos , Ratones , Ratones Noqueados , Células-Madre Neurales/metabolismo
6.
FASEB J ; 29(4): 1480-92, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25636741

RESUMEN

We studied potential changes in the subventricular zone (SVZ) stem cell niche of the senescence-accelerated mouse prone-8 (SAM-P8) aging model. Bromodeoxyuridine (BrdU) assays with longtime survival revealed a lower number of label-retaining stem cells in the SAM-P8 SVZ compared with the SAM-Resistant 1 (SAM-R1) control strain. We also found that in SAM-P8 niche signaling is attenuated and the stem cell pool is less responsive to the self-renewal niche factor pigmented epithelium-derived factor (PEDF). Protein analysis demonstrated stable amounts of the PEDF ligand in the SAM-P8 SVZ niche; however, SAM-P8 stem cells present a significant expression decrease of patatin-like phospholipase domain containing 2, a receptor for PEDF (PNPLA2-PEDF) receptor, but not of laminin receptor (LR), a receptor for PEDF (LR-PEDF) receptor. We observed changes in self-renewal related genes (hairy and enhancer of split 1 (Hes1), hairy and enhancer of split 1 (Hes5), Sox2] and report that although these genes are down-regulated in SAM-P8, differentiation genes (Pax6) are up-regulated and neurogenesis is increased. Finally, sheltering mammalian telomere complexes might be also involved given a down-regulation of telomeric repeat binding factor 1 (Terf1) expression was observed in SAM-P8 at young age periods. Differences between these 2 models, SAM-P8 and SAM-R1 controls, have been previously detected at more advanced ages. We now describe alterations in the PEDF signaling pathway and stem cell self-renewal at a very young age, which could be involved in the premature senescence observed in the SAM-P8 model.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/patología , Proteínas del Ojo/metabolismo , Ventrículos Laterales/metabolismo , Ventrículos Laterales/patología , Factores de Crecimiento Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Serpinas/metabolismo , Envejecimiento/genética , Animales , Bromodesoxiuridina/metabolismo , Recuento de Células , Proteínas del Ojo/genética , Ratones , Modelos Animales , Modelos Neurológicos , Factores de Crecimiento Nervioso/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Neuropéptido/genética , Receptores de Neuropéptido/metabolismo , Serpinas/genética , Transducción de Señal , Nicho de Células Madre
7.
Proc Natl Acad Sci U S A ; 108(10): 4194-9, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21325059

RESUMEN

The aggregation of proteins into oligomers and amyloid fibrils is characteristic of several neurodegenerative diseases, including Parkinson disease (PD). In PD, the process of aggregation of α-synuclein (α-syn) from monomers, via oligomeric intermediates, into amyloid fibrils is considered the disease-causative toxic mechanism. We developed α-syn mutants that promote oligomer or fibril formation and tested the toxicity of these mutants by using a rat lentivirus system to investigate loss of dopaminergic neurons in the substantia nigra. The most severe dopaminergic loss in the substantia nigra is observed in animals with the α-syn variants that form oligomers (i.e., E57K and E35K), whereas the α-syn variants that form fibrils very quickly are less toxic. We show that α-syn oligomers are toxic in vivo and that α-syn oligomers might interact with and potentially disrupt membranes.


Asunto(s)
Biopolímeros/toxicidad , alfa-Sinucleína/toxicidad , Animales , Encéfalo/metabolismo , Lentivirus/genética , Ratas , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
8.
Stem Cells ; 30(12): 2796-809, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22987443

RESUMEN

Adult neural stem cells (NSCs) located in the subventricular zone (SVZ) persistently produce new neurons destined to the olfactory bulb (OB). Recent research suggests that the OB is also a source of NSCs that remains largely unexplored. Using single/dual-labeling procedures, we address the existence of NSCs in the innermost layers of the OB. In vivo, these cells are more quiescent that their SVZ counterparts, but after in vitro expansion, they behave similarly. Self-renewal and proliferation assays in co-culture with niche astrocytes indicate that OB-glia restricts NSC activity whereas SVZ-glia has the opposite effect. Gene expression profiling identifies WNT7A as a key SVZ-glial factor lacking in OB-glia that enhances self-renewal, thereby improving the propagation of OB-NSC cultures. These data demonstrate that region-specific glial factors account for in vivo differences in NSC activity and point to WNT7A as a tool that may be instrumental for the NSC expansion phase that precedes grafting.


Asunto(s)
Astrocitos/citología , Células-Madre Neurales/citología , Bulbo Olfatorio/citología , Proteínas Wnt/metabolismo , Animales , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Humanos , Ratones , Proteínas Wnt/genética
9.
Nat Commun ; 14(1): 7541, 2023 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-38001081

RESUMEN

Neurogenesis in the adult mammalian brain relies on the lifelong persistence of quiescent neural stem cell (NSC) reservoirs. Little is known about the mechanisms that lead to the initial establishment of quiescence, the main hallmark of adult stem cells, during development. Here we show that protein aggregates and autophagy machinery components accumulate in developmental radial glia-like NSCs as they enter quiescence and that pharmacological or genetic blockade of autophagy disrupts quiescence acquisition and maintenance. Conversely, increasing autophagy through AMPK/ULK1 activation instructs the acquisition of the quiescent state without affecting BMP signaling, a gatekeeper of NSC quiescence during adulthood. Selective ablation of Atg7, a critical gene for autophagosome formation, in radial glia-like NSCs at early and late postnatal stages compromises the initial acquisition and maintenance of quiescence during the formation of the hippocampal dentate gyrus NSC niche. Therefore, we demonstrate that autophagy is cell-intrinsically required to establish NSC quiescence during hippocampal development. Our results uncover an important role of autophagy in the transition of developmental NSCs into their dormant adult form, paving the way for studies directed at further understanding the mechanisms of stem cell niche formation and maintenance in the mammalian brain.


Asunto(s)
Células Madre Adultas , Células-Madre Neurales , Animales , Neurogénesis/genética , Hipocampo , División Celular , Autofagia/genética , Células-Madre Neurales/metabolismo , Células Madre Adultas/metabolismo , Mamíferos
10.
Front Mol Neurosci ; 16: 1237458, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37900943

RESUMEN

Degeneration of basal forebrain cholinergic neurons (BFCNs) is a hallmark of Alzheimer's disease (AD). However, few mouse models of AD recapitulate the neurodegeneration of the cholinergic system. The p75 neurotrophin receptor, p75NTR, has been associated with the degeneration of BFCNs in AD. The senescence-accelerated mouse prone number 8 (SAMP8) is a well-accepted model of accelerated and pathological aging. To gain a better understanding of the role of p75NTR in the basal forebrain during aging, we generated a new mouse line, the SAMP8-p75exonIII-/-. Deletion of p75NTR in the SAMP8 background induces an increase in the number of BFCNs at birth, followed by a rapid decline during aging compared to the C57/BL6 background. This decrease in the number of BFCNs correlates with a worsening in the Y-maze memory test at 6 months in the SAMP8-p75exonIII-/-. We found that SAMP8-p75exonIII-/- and C57/BL6-p75exonIII-/- mice expressed constitutively a short isoform of p75NTR that correlates with an upregulation of the protein levels of SREBP2 and its targets, HMGCR and LDLR, in the BF of both SAMP8-p75exonIII-/- and C57/BL6-p75exonIII-/- mice. As the neurodegeneration of the cholinergic system and the dysregulation of cholesterol metabolism are implicated in AD, we postulate that the generated SAMP8-p75exonIII-/- mouse strain might constitute a good model to study long-term cholinergic neurodegeneration in the CNS. In addition, our results support the role of p75NTR signaling in cholesterol biosynthesis regulation.

11.
Front Cell Dev Biol ; 10: 912319, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35938168

RESUMEN

Stem cells in adult mammalian tissues are held in a reversible resting state, known as quiescence, for prolonged periods of time. Recent studies have greatly increased our understanding of the epigenetic and transcriptional landscapes that underlie stem cell quiescence. However, the transcription factor code that actively maintains the quiescence program remains poorly defined. Similarly, alternative splicing events affecting transcription factors in stem cell quiescence have been overlooked. Here we show that the transcription factor T-cell factor/lymphoid enhancer factor LEF1, a central player in canonical ß-catenin-dependent Wnt signalling, undergoes alternative splicing and switches isoforms in quiescent neural stem cells. We found that active ß-catenin and its partner LEF1 accumulated in quiescent hippocampal neural stem and progenitor cell (Q-NSPC) cultures. Accordingly, Q-NSPCs showed enhanced TCF/LEF1-driven transcription and a basal Wnt activity that conferred a functional advantage to the cultured cells in a Wnt-dependent assay. At a mechanistic level, we found a fine regulation of Lef1 gene expression. The coordinate upregulation of Lef1 transcription and retention of alternative spliced exon 6 (E6) led to the accumulation of a full-length protein isoform (LEF1-FL) that displayed increased stability in the quiescent state. Prospectively isolated GLAST + cells from the postnatal hippocampus also underwent E6 retention at the time quiescence is established in vivo. Interestingly, LEF1 motif was enriched in quiescence-associated enhancers of genes upregulated in Q-NSPCs and quiescence-related NFIX transcription factor motifs flanked the LEF1 binding sites. We further show that LEF1 interacts with NFIX and identify putative LEF1/NFIX targets. Together, our results uncover an unexpected role for LEF1 in gene regulation in quiescent NSPCs, and highlight alternative splicing as a post-transcriptional regulatory mechanism in the transition from stem cell activation to quiescence.

12.
Cell Rep ; 38(5): 110313, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108528

RESUMEN

The adult neurogenic niche in the hippocampus is maintained through activation of reversibly quiescent neural stem cells (NSCs) with radial glia-like morphology (RGLs). Here, we show that the expression of SoxD transcription factors Sox5 and Sox6 is enriched in activated RGLs. Using inducible deletion of Sox5 or Sox6 in the adult mouse brain, we show that both genes are required for RGL activation and the generation of new neurons. Conversely, Sox5 overexpression in cultured NSCs interferes with entry in quiescence. Mechanistically, expression of the proneural protein Ascl1 (a key RGL regulator) is severely downregulated in SoxD-deficient RGLs, and Ascl1 transcription relies on conserved Sox motifs. Additionally, loss of Sox5 hinders the RGL activation driven by neurogenic stimuli such as environmental enrichment. Altogether, our data suggest that SoxD genes are key mediators in the transition of adult RGLs from quiescence to an activated mitotic state under physiological situations.


Asunto(s)
Células Madre Adultas/metabolismo , Células-Madre Neurales/metabolismo , Factores de Transcripción SOXD/metabolismo , Animales , Diferenciación Celular/fisiología , Hipocampo/metabolismo , Ratones Transgénicos , Neurogénesis/fisiología , Factores de Transcripción SOXD/genética , Factores de Transcripción/metabolismo
13.
Nature ; 437(7063): 1370-5, 2005 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-16251967

RESUMEN

The generation of new neurons from neural stem cells is restricted to two regions of the adult mammalian central nervous system: the subventricular zone of the lateral ventricle, and the subgranular zone of the hippocampal dentate gyrus. In both regions, signals provided by the microenvironment regulate the maintenance, proliferation and neuronal fate commitment of the local stem cell population. The identity of these signals is largely unknown. Here we show that adult hippocampal stem/progenitor cells (AHPs) express receptors and signalling components for Wnt proteins, which are key regulators of neural stem cell behaviour in embryonic development. We also show that the Wnt/beta-catenin pathway is active and that Wnt3 is expressed in the hippocampal neurogenic niche. Overexpression of Wnt3 is sufficient to increase neurogenesis from AHPs in vitro and in vivo. By contrast, blockade of Wnt signalling reduces neurogenesis from AHPs in vitro and abolishes neurogenesis almost completely in vivo. Our data show that Wnt signalling is a principal regulator of adult hippocampal neurogenesis and provide evidence that Wnt proteins have a role in adult hippocampal function.


Asunto(s)
Hipocampo/citología , Hipocampo/metabolismo , Neuronas/citología , Neuronas/metabolismo , Transducción de Señal , Envejecimiento/fisiología , Animales , Astrocitos/citología , Astrocitos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Células Madre/citología , Células Madre/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt3
14.
J Neurosci ; 29(46): 14394-407, 2009 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-19923274

RESUMEN

Proliferation in the subependymal zone (SEZ) and neurogenesis in the olfactory bulb decline in the forebrain of telomerase-deficient mice. The present work reveals additional effects of telomere shortening on neuronal differentiation, as adult multipotent progenitors with critically short telomeres yield reduced numbers of neurons that, furthermore, exhibit underdeveloped neuritic arbors. Genetic data indicate that the tumor suppressor protein p53 not only mediates the adverse effects of telomere attrition on proliferation and self-renewal but it is also involved in preventing normal neuronal differentiation of adult progenitors with dysfunctional telomeres. Interestingly, progenitor cells with short telomeres obtained from fetal brains do not exhibit any replicative defects but also fail to acquire a fully mature neuritic arbor, demonstrating cell cycle-independent effects of telomeres on neuronal differentiation. The negative effect of p53 on neuritogenesis is mechanistically linked to its cooperation with the Notch pathway in the upregulation of small GTPase RhoA kinases, Rock1 and Rock2, suggesting a potential link between DNA damage and the Notch signaling pathway in the control of neuritogenesis. We also show that telomerase expression is downregulated in the SEZ of aging mice leading to telomere length reductions in neurosphere-forming cells and deficient neurogenesis and neuritogenesis. Our results suggest that age-related deficits could be caused partly by dysfunctional telomeres and demonstrate that p53 is a central modulator of adult neurogenesis, regulating both the production and differentiation of postnatally generated olfactory neurons.


Asunto(s)
Diferenciación Celular , Neuritas/patología , Neurogénesis , Células Madre/patología , Telómero/patología , Envejecimiento/genética , Envejecimiento/patología , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Células Cultivadas , Feto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuritas/enzimología , Neurogénesis/genética , Neuronas , Receptores Notch/fisiología , Transducción de Señal/genética , Células Madre/enzimología , Telomerasa/deficiencia , Telomerasa/genética , Telómero/enzimología , Proteína p53 Supresora de Tumor/fisiología , Quinasas Asociadas a rho/biosíntesis , Quinasas Asociadas a rho/metabolismo
15.
Nat Neurosci ; 9(3): 331-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16491078

RESUMEN

Adult stem cells are characterized by self-renewal and multilineage differentiation, and these properties seem to be regulated by signals from adjacent differentiated cell types and by extracellular matrix molecules, which collectively define the stem cell "niche." Self-renewal is essential for the lifelong persistence of stem cells, but its regulation is poorly understood. In the mammalian brain, neurogenesis persists in two germinal areas, the subventricular zone (SVZ) and the hippocampus, where continuous postnatal neuronal production seems to be supported by neural stem cells (NSCs). Here we show that pigment epithelium-derived factor (PEDF) is secreted by components of the murine SVZ and promotes self-renewal of adult NSCs in vitro. In addition, intraventricular PEDF infusion activated slowly dividing stem cells, whereas a blockade of endogenous PEDF decreased their cycling. These data demonstrate that PEDF is a niche-derived regulator of adult NSCs and provide evidence for a role for PEDF protein in NSC maintenance.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Proteínas del Ojo/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Serpinas/metabolismo , Células Madre/metabolismo , Telencéfalo/metabolismo , Animales , Células COS , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Células Cultivadas , Chlorocebus aethiops , Endotelio Vascular/metabolismo , Epéndimo/citología , Epéndimo/efectos de los fármacos , Epéndimo/metabolismo , Proteínas del Ojo/farmacología , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Inyecciones Intraventriculares , Ventrículos Laterales/citología , Ventrículos Laterales/metabolismo , Ratones , Factores de Crecimiento Nervioso/farmacología , Plasticidad Neuronal/efectos de los fármacos , Neuronas/citología , Serpinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Madre/efectos de los fármacos , Telencéfalo/citología , Telencéfalo/efectos de los fármacos
16.
Front Cell Dev Biol ; 8: 533, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32695783

RESUMEN

The human brain is composed of billions of cells, including neurons and glia, with an undetermined number of subtypes. During the embryonic and early postnatal stages, the vast majority of these cells are generated from neural progenitors and stem cells located in all regions of the neural tube. A smaller number of neurons will continue to be generated throughout our lives, in localized neurogenic zones, mainly confined at least in rodents to the subependymal zone of the lateral ventricles and the subgranular zone of the hippocampal dentate gyrus. During neurogenesis, a combination of extrinsic cues interacting with temporal and regional intrinsic programs are thought to be critical for increasing neuronal diversity, but their underlying mechanisms need further elucidation. In this review, we discuss the recent findings in Drosophila and mammals on the types of cell division and cell interactions used by neural progenitors and stem cells to sustain neurogenesis, and how they are influenced by glia.

17.
Front Cell Dev Biol ; 8: 686, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32903900

RESUMEN

[This corrects the article DOI: 10.3389/fcell.2020.00533.].

18.
Stem Cells ; 26(7): 1891-900, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18436867

RESUMEN

Increasing evidence suggests that alpha-chemokines serve several important functions in the nervous system, including regulation of neuroimmune responses, neurotransmission, neuronal survival, and central nervous system development. In this study, we first examined the function of two alpha-chemokines, chemokine ligand (CXCL) 6 and CXCL8, and their receptors, CXCR1 and CXCR2, in the developing rat ventral midbrain (VM). We found that CXCR2 and CXCL6 are regulated during VM development and that CXCL6 promotes the differentiation of nurr77-related receptor (Nurr1)+ precursors into dopaminergic (DA) neurons in vitro. Intriguingly, CXCL8, a ligand expressed only in Homo sapiens, enhanced progenitor cell division, neurogenesis, and tyrosine hydroxylase-positive (TH+) cell number in rodent precursor and neurosphere cultures. CXCL1, the murine ortholog of CXCL8, was developmentally regulated in the VM and exhibited activities similar but not identical to those of CXCL8. TH+ cells derived from chemokine-treated VM neurospheres coexpressed Nurr1 and VMAT and were functionally active, as shown by calcium (Ca(2+)) fluxes in response to AMPA. In conclusion, our data demonstrate that CXCL1, CXCL6, and CXCL8 increase the number of DA neurons in VM precursor and neurosphere cultures by diverse mechanisms. Thus, alpha-chemokines may find an application in the preparation of cells for drug development or Parkinson's disease cell replacement therapy.


Asunto(s)
Quimiocinas/biosíntesis , Dopamina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mesencéfalo/embriología , Mesencéfalo/metabolismo , Neuronas/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Femenino , Interleucina-8/metabolismo , Masculino , Ratones , Ratas , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/metabolismo , Células Madre/citología
19.
Exp Cell Res ; 314(10): 2123-30, 2008 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-18420193

RESUMEN

beta-chemokines are secreted factors that regulate diverse functions in the adult brain, such as neuro-immune responses and neurotransmission, but their function in the developing brain is largely unknown. We recently found that the orphan nuclear receptor, Nurr1, up regulates CCL2 and CCL7 in neural stem cells, suggesting a possible function of beta-chemokines in midbrain development. Here we report that two beta-chemokines, CCL2 and CCL7, and two of their receptors, CCR1 and CCR2, are expressed and developmentally regulated in the ventral midbrain (VM). Moreover, we found that the expression of CCL7 was down regulated in the Nurr1 knockout mice, linking CCL7 to dopamine (DA) neuron development. When the function of CCL2 and CCL7 was examined, we found that they selectively enhanced the differentiation of Nurr1+ precursors into DA neurons, but not their survival or progenitor proliferation in primary precursor cultures. Moreover, both CCL2 and CCL7 promoted neuritogenesis in midbrain DA neuron cultures. Thus, our results show for the first time a function of beta-chemokines in the developing brain and identify beta-chemokines as novel class of pro-differentiation factors for midbrain DA neurons. These data also suggest that beta-chemokines may become useful tools to enhance the differentiation of DA cell preparations for cell replacement therapy and drug discovery in Parkinson's disease (PD).


Asunto(s)
Diferenciación Celular/fisiología , Quimiocina CCL2/metabolismo , Quimiocina CCL7/metabolismo , Dopamina/metabolismo , Mesencéfalo/citología , Neuronas/metabolismo , Células Madre/fisiología , Animales , Proliferación Celular , Supervivencia Celular , Quimiocina CCL2/genética , Quimiocina CCL7/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Masculino , Mesencéfalo/embriología , Mesencéfalo/metabolismo , Ratones , Ratones Noqueados , Neuronas/citología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Ratas , Ratas Sprague-Dawley , Receptores CCR1/genética , Receptores CCR1/metabolismo , Receptores CCR2/genética , Receptores CCR2/metabolismo , Células Madre/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Front Cell Dev Biol ; 7: 96, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31214589

RESUMEN

The generation of new neurons is a lifelong process in many vertebrate species that provides an extra level of plasticity to several brain circuits. Frequently, neurogenesis in the adult brain is considered a continuation of earlier developmental processes as it relies in the persistence of neural stem cells, similar to radial glia, known as radial glia-like cells (RGLs). However, adult RGLs are not just leftovers of progenitors that remain in hidden niches in the brain after development has finished. Rather, they seem to be specified and set aside at specific times and places during embryonic and postnatal development. The adult RGLs present several cellular and molecular properties that differ from those observed in developmental radial glial cells such as an extended cell cycle length, acquisition of a quiescence state, a more restricted multipotency and distinct transcriptomic programs underlying those cellular processes. In this minireview, we will discuss the recent attempts to determine how, when and where are the adult RGLs specified.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA