Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Adv Exp Med Biol ; 1041: 81-118, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29204830

RESUMO

The cells that build the nervous system, either this is a small network of ganglia or a complicated primate brain, are called neural stem and progenitor cells. Even though the very primitive and the very recent neural stem cells (NSCs) share common basic characteristics that are hard-wired within their character, such as the expression of transcription factors of the SoxB family, their capacity to give rise to extremely different neural tissues depends significantly on instructions from the microenvironment. In this chapter we explore the nature of the NSC microenvironment, looking through evolution, embryonic development, maturity and even disease. Experimental work undertaken over the last 20 years has revealed exciting insight into the NSC microcosmos. NSCs are very capable in producing their own extracellular matrix and in regulating their behaviour in an autocrine and paracrine manner. Nevertheless, accumulating evidence indicates an important role for the vasculature, especially within the NSC niches of the postnatal brain; while novel results reveal direct links between the metabolic state of the organism and the function of NSCs.


Assuntos
Matriz Extracelular/fisiologia , Microvasos/fisiologia , Células-Tronco Neurais/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/embriologia , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/fisiopatologia , Diferenciação Celular/fisiologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Humanos , Células-Tronco Neurais/citologia , Neurônios/fisiologia
2.
Cells ; 13(8)2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38667283

RESUMO

Astrocytes and ependymal cells have been reported to be able to switch from a mature cell identity towards that of a neural stem/progenitor cell. Astrocytes are widely scattered in the brain where they exert multiple functions and are routinely targeted for in vitro and in vivo reprogramming. Ependymal cells serve more specialized functions, lining the ventricles and the central canal, and are multiciliated, epithelial-like cells that, in the spinal cord, act as bi-potent progenitors in response to injury. Here, we isolate or generate ependymal cells and post-mitotic astrocytes, respectively, from the lateral ventricles of the mouse brain and we investigate their capacity to reverse towards a progenitor-like identity in culture. Inhibition of the GSK3 and TGFß pathways facilitates the switch of mature astrocytes to Sox2-expressing, mitotic cells that generate oligodendrocytes. Although this medium allows for the expansion of quiescent NSCs, isolated from live rats by "milking of the brain", it does not fully reverse astrocytes towards the bona fide NSC identity; this is a failure correlated with a concomitant lack of neurogenic activity. Ependymal cells could be induced to enter mitosis either via exposure to neuraminidase-dependent stress or by culturing them in the presence of FGF2 and EGF. Overall, our data confirm that astrocytes and ependymal cells retain a high capacity to reverse to a progenitor identity and set up a simple and highly controlled platform for the elucidation of the molecular mechanisms that regulate this reversal.


Assuntos
Astrócitos , Epêndima , Fenótipo , Animais , Astrócitos/metabolismo , Astrócitos/citologia , Epêndima/citologia , Epêndima/metabolismo , Camundongos , Células Cultivadas , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Diferenciação Celular , Encéfalo/citologia , Encéfalo/metabolismo , Ratos , Fatores de Transcrição SOXB1/metabolismo , Camundongos Endogâmicos C57BL , Mitose , Quinase 3 da Glicogênio Sintase/metabolismo , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Animais Recém-Nascidos
3.
J Vis Exp ; (204)2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38407295

RESUMO

Tissue-specific neural stem cells (NSCs) remain active in the mammalian postnatal brain. They reside in specialized niches, where they generate new neurons and glia. One such niche is the subependymal zone (SEZ; also called the ventricular-subventricular zone), which is located across the lateral walls of the lateral ventricles, adjacent to the ependymal cell layer. Oligodendrocyte progenitor cells (OPCs) are abundantly distributed throughout the central nervous system, constituting a pool of proliferative progenitor cells that can generate oligodendrocytes. Both NSCs and OPCs exhibit self-renewal potential and quiescence/activation cycles. Due to their location, the isolation and experimental investigation of these cells is performed postmortem. Here, we describe in detail "brain milking", a method for the isolation of NSCs and OPCs, amongst other cells, from live animals. This is a two-step protocol designed for use in rodents and tested in rats. First, cells are "released" from the tissue via stereotaxic intracerebroventricular (i.c.v.) injection of a "release cocktail". The main components are neuraminidase, which targets ependymal cells and induces ventricular wall denudation, an integrin-ß1-blocking antibody, and fibroblast growth factor-2. At a second "collection" step, liquid biopsies of cerebrospinal fluid are performed from the cisterna magna, in anesthetized rats without the need of an incision. Results presented here show that isolated cells retain their endogenous profile and that NSCs of the SEZ preserve their quiescence. The denudation of the ependymal layer is restricted to the anatomical level of injection and the protocol (release and collection) is tolerated well  by the animals. This novel approach paves the way for performing longitudinal studies of endogenous neurogenesis and gliogenesis in experimental animals.


Assuntos
Células-Tronco Neurais , Células Precursoras de Oligodendrócitos , Ratos , Animais , Encéfalo , Sistema Nervoso Central , Neuroglia , Mamíferos
4.
MicroPubl Biol ; 20242024.
Artigo em Inglês | MEDLINE | ID: mdl-38550606

RESUMO

Alpha-synuclein plays a pivotal role in Parkinson's disease (PD) pathogenesis, with α-synuclein aggregates/oligomers being identified as toxic species and phosphorylation at Serine 129 promoting aggregation/oligomerization. We investigated the biochemical profile of α-synuclein in the "weaver" mouse, a genetic PD model. Our results revealed increased Serine 129 phosphorylation in the midbrain, striatum, and cortex at a phase of established dopaminergic degeneration on postnatal day 100. These results indicate α-synuclein pathology already at this stage and the potential for age-related progress. Our findings confirm that the "weaver" mouse is an invaluable genetic model to study α-synuclein pathogenesis during PD progression.

5.
J Cell Sci ; 123(Pt 10): 1613-22, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20445012

RESUMO

The expression of adhesion molecules by stem cells within their niches is well described, but what is their function? A conventional view is that these adhesion molecules simply retain stem cells in the niche and thereby maintain its architecture and shape. Here, we review recent literature showing that this is but one of their roles, and that they have essential functions in all aspects of the stem cell-niche interaction--retention, division and exit. We also highlight from this literature evidence supporting a simple model whereby the regulation of centrosome positioning and spindle angle is regulated by both cadherins and integrins, and the differential activity of these two adhesion molecules enables the fundamental stem cell property of switching between asymmetrical and symmetrical divisions.


Assuntos
Caderinas/metabolismo , Integrinas/metabolismo , Nicho de Células-Tronco/metabolismo , Células-Tronco/metabolismo , Animais , Divisão do Núcleo Celular , Humanos , Modelos Biológicos , Nicho de Células-Tronco/patologia , Células-Tronco/patologia
6.
Neural Regen Res ; 17(12): 2637-2642, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35662194

RESUMO

Parkinson's disease is the second most common neurodegenerative disease and has currently no effective treatment, one that would be able to stop or reverse the loss of dopaminergic neurons in the substantia nigra pars compacta. In addition, Parkinson's disease diagnosis is typically done when a significant percentage of the dopaminergic neurons is already lost. In neurodegenerative disorders, some therapeutic strategies could be effective only at inhibiting further degeneration; on the other hand, cell replacement therapies aim at replacing lost neurons, an approach that would be ideal for the treatment of Parkinson's disease. Many cell replacement therapies have been tested since the 1970s in the field of Parkinson's disease; however, there are still significant limitations prohibiting a successful clinical application. From the first fetal midbrain intrastriatal graft to the most recent conversion of astrocytes into dopaminergic neurons, we have gained equally, significant insights and questions still looking for an answer. This review aims to summarize the main milestones in cell replacement approaches against Parkinson's disease. By focusing on achievements and failures, as well as on the additional research steps needed, we aim to provide perspective on how future cell replacement therapies treats Parkinson's disease.

7.
Cells ; 11(11)2022 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-35681436

RESUMO

Two main stem cell pools exist in the postnatal mammalian brain that, although they share some "stemness" properties, also exhibit significant differences. Multipotent neural stem cells survive within specialized microenvironments, called niches, and they are vulnerable to ageing. Oligodendroglial lineage-restricted progenitor cells are widely distributed in the brain parenchyma and are more resistant to the effects of ageing. Here, we create polymorphic neural stem cell cultures and allow cells to progress towards the neuronal and the oligodendroglial lineage. We show that the divergence of cell fate is accompanied by a divergence in the properties of progenitors, which reflects their adaptation to life in the niche or the parenchyma. Neurogenesis shows significant spatial restrictions and a dependence on laminin, a major niche component, while oligodendrogenesis shows none of these constraints. Furthermore, the blocking of integrin-ß1 leads to opposing effects, reducing neurogenesis and enhancing oligodendrogenesis. Therefore, polymorphic neural stem cell assays can be used to investigate the divergence of postnatal brain stem cells and also to predict the in vivo effects of potential therapeutic molecules targeting stem and progenitor cells, as we do for the microneurotrophin BNN-20.


Assuntos
Células-Tronco Neurais , Animais , Encéfalo , Mamíferos , Neurogênese/fisiologia , Neurônios , Oligodendroglia
8.
Int J Dev Biol ; 66(1-2-3): 51-58, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35238392

RESUMO

In the postnatal mammalian brain, neurogenic activity is retained in anatomically restricted areas, driven by pools of Neural Stem Cells (NSCs). These cells and their progeny have been studied intensively as potential targets for regenerative treatments, aiming at either their in situ manipulation or their use as sources of cells for transplantation-based strategies. Although their full identity, heterogeneity and differentiation potential remain elusive, due to the absence of specific cell-type markers, our knowledge of their properties is constantly expanding. Here, we focus on the NSC niche that is located at the Subependymal Zone (SEZ/ also known as Subventricular Zone) of the lateral ventricles of the brain. We review, summarize and explain the different faces of the NSC, as they have been described, using a wide range of experimental approaches, over a time-frame of three decades: the primitive, definitive, quiescent or activated NSC. We also review the growing evidence of the existence of latent NSCs outside of niches, in the brain parenchyma, that constitute promising new therapeutic targets, complemented by the novel technologies of in vivo cell reprogramming.


Assuntos
Células-Tronco Neurais , Nicho de Células-Tronco , Animais , Encéfalo , Ventrículos Laterais , Mamíferos , Neurogênese
9.
J Neurosci ; 30(29): 9771-81, 2010 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-20660259

RESUMO

The subependymal zone (SEZ) of the lateral ventricles is one of the areas of the adult brain where new neurons are continuously generated from neural stem cells (NSCs), via rapidly dividing precursors. This neurogenic niche is a complex cellular and extracellular microenvironment, highly vascularized compared to non-neurogenic periventricular areas, within which NSCs and precursors exhibit distinct behavior. Here, we investigate the possible mechanisms by which extracellular matrix molecules and their receptors might regulate this differential behavior. We show that NSCs and precursors proceed through mitosis in the same domains within the SEZ of adult male mice--albeit with NSCs nearer ependymal cells--and that distance from the ventricle is a stronger limiting factor for neurogenic activity than distance from blood vessels. Furthermore, we show that NSCs and precursors are embedded in a laminin-rich extracellular matrix, to which they can both contribute. Importantly, they express differential levels of extracellular matrix receptors, with NSCs expressing low levels of alpha6beta1 integrin, syndecan-1, and lutheran, and in vivo blocking of beta1 integrin selectively induced the proliferation and ectopic migration of precursors. Finally, when NSCs are activated to reconstitute the niche after depletion of precursors, expression of laminin receptors is upregulated. These results indicate that the distinct behavior of adult NSCs and precursors is not necessarily regulated via exposure to differential extracellular signals, but rather via intrinsic regulation of their interaction with their microenvironment.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Epêndima/citologia , Epêndima/metabolismo , Matriz Extracelular/metabolismo , Receptores de Laminina/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Movimento Celular/fisiologia , Proliferação de Células , Integrina beta1/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Mitose
10.
Stem Cell Res Ther ; 12(1): 335, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34112234

RESUMO

BACKGROUND: Loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) underlines much of the pathology of Parkinson's disease (PD), but the existence of an endogenous neurogenic system that could be targeted as a therapeutic strategy has been controversial. BNN-20 is a synthetic, BDNF-mimicking, microneurotrophin that we previously showed to exhibit a pleiotropic neuroprotective effect on the dopaminergic neurons of the SNpc in the "weaver" mouse model of PD. Here, we assessed its potential effects on neurogenesis. METHODS: We quantified total numbers of dopaminergic neurons in the SNpc of wild-type and "weaver" mice, with or without administration of BNN-20, and we employed BrdU labelling and intracerebroventricular injections of DiI to evaluate the existence of dopaminergic neurogenesis in the SNpc and to assess the origin of newborn dopaminergic neurons. The in vivo experiments were complemented by in vitro proliferation/differentiation assays of adult neural stem cells (NSCs) isolated from the substantia nigra and the subependymal zone (SEZ) stem cell niche to further characterize the effects of BNN-20. RESULTS: Our analysis revealed the existence of a low-rate turnover of dopaminergic neurons in the normal SNpc and showed, using three independent lines of experiments (stereologic cell counts, BrdU and DiI tracing), that the administration of BNN-20 leads to increased neurogenesis in the SNpc and to partial reversal of dopaminergic cell loss. The newly born dopaminergic neurons, that are partially originated from the SEZ, follow the typical nigral maturation pathway, expressing the transcription factor FoxA2. Importantly, the pro-cytogenic effects of BNN-20 were very strong in the SNpc, but were absent in other brain areas such as the cortex or the stem cell niche of the hippocampus. Moreover, although the in vitro assays showed that BNN-20 enhances the differentiation of NSCs towards glia and neurons, its in vivo administration stimulated only neurogenesis. CONCLUSIONS: Our results demonstrate the existence of a neurogenic system in the SNpc that can be manipulated in order to regenerate the depleted dopaminergic cell population in the "weaver" PD mouse model. Microneurotrophin BNN-20 emerges as an excellent candidate for future PD cell replacement therapies, due to its area-specific, pro-neurogenic effects.


Assuntos
Neurogênese , Substância Negra , Animais , Dopamina , Neurônios Dopaminérgicos , Homeostase , Camundongos
11.
Stem Cell Reports ; 16(10): 2534-2547, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34560001

RESUMO

Postnatal brain neural stem and progenitor cells (NSPCs) cluster in anatomically inaccessible stem cell niches, such as the subependymal zone (SEZ). Here, we describe a method for the isolation of NSPCs from live animals, which we term "milking." The intracerebroventricular injection of a release cocktail, containing neuraminidase, integrin-ß1-blocking antibody, and fibroblast growth factor 2, induces the controlled flow of NSPCs in the cerebrospinal fluid, where they are collected via liquid biopsies. Isolated cells retain key in vivo self-renewal properties and their cell-type profile reflects the cell composition of their source area, while the function of the niche is sustained even 8 months post-milking. By changing the target area more caudally, we also isolate oligodendrocyte progenitor cells (OPCs) from the corpus callosum. This novel approach for sampling NSPCs and OPCs paves the way for performing longitudinal studies in experimental animals, for more in vivo relevant cell culture assays, and for future clinical neuro-regenerative applications.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Neurais/metabolismo , Células Precursoras de Oligodendrócitos/metabolismo , Animais , Encéfalo , Diferenciação Celular , Líquido Cefalorraquidiano , Corpo Caloso , Humanos , Biópsia Líquida , Masculino , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Ratos Wistar , Nicho de Células-Tronco
12.
Brain ; 132(Pt 11): 2909-21, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19773354

RESUMO

The mammalian brain is a remarkably complex organ comprising millions of neurons, glia and various other cell types. Its impressive cytoarchitecture led to the long standing belief that it is a structurally static organ and thus very sensitive to injury. However, an area of striking structural flexibility has been recently described at the centre of the brain. It is the subependymal zone of the lateral wall of the lateral ventricles. The subependymal zone--like a beating heart--continuously sends new cells to different areas of the brain: neurons to the olfactory bulbs and glial cells to the cortex and the corpus callosum. Interestingly, the generation and flow of cells changes in response to signals from anatomically remote areas of the brain or even from the external environment of the organism, therefore indicating that subependymal neurogenesis--as a system--is integrated in the overall homeostatic function of the brain. In this review, it will be attempted to describe the fundamental structural and functional characteristics of the subependymal neurogenic niche and to summarize the available evidence regarding its plasticity. Special focus is given on issues such as whether adult neural stem cells are activated after neurodegeneration, whether defects in neurogenesis contribute to neuropathological conditions and whether monitoring changes in neurogenic activity can have a diagnostic value.


Assuntos
Encéfalo/anatomia & histologia , Encéfalo/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Nicho de Células-Tronco , Animais , Encéfalo/patologia , Linhagem da Célula , Movimento Celular/fisiologia , Meio Ambiente , Epêndima/citologia , Epêndima/fisiologia , Degeneração Neural/patologia , Plasticidade Neuronal/fisiologia , Neurônios/citologia , Transdução de Sinais/fisiologia , Nicho de Células-Tronco/anatomia & histologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/fisiologia
13.
J Neurosci ; 27(51): 13991-6, 2007 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-18094237

RESUMO

The subependymal zone (SEZ) of the lateral ventricles of the adult mouse brain hosts neurogenesis from a neural stem cell population with the morphology of astrocytes (termed type-B cells). Tenascin-C is a large extracellular matrix glycoprotein present in the SEZ that has been shown to regulate the development of embryonic neural stem cells and the proliferation and migration of early postnatal neural precursors. Here we show that tenascin-C is produced by type-B cells and forms a layer between SEZ and the adjacent striatum. Tenascin-C deficiency resulted in minor structural differences in and around the SEZ. However, the numbers of neural stem cells and their progeny remained unaffected, as did their regeneration after depletion of mitotic cells using the antimitotic drug cytosine-beta-D-arabinofuranoside. Our results reveal a remarkable ability of the adult neural stem cell niche to retain proper function even after the removal of major extracellular matrix molecules.


Assuntos
Epêndima/fisiologia , Ventrículos Laterais/fisiologia , Regeneração Nervosa/fisiologia , Tenascina/deficiência , Fatores Etários , Animais , Astrócitos/metabolismo , Astrócitos/fisiologia , Corpo Estriado/citologia , Corpo Estriado/metabolismo , Corpo Estriado/fisiologia , Epêndima/citologia , Epêndima/metabolismo , Ventrículos Laterais/citologia , Ventrículos Laterais/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco/citologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia
14.
PLoS Biol ; 3(6): e186, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15882093

RESUMO

Little is known about the molecular mechanisms and intrinsic factors that are responsible for the emergence of neuronal subtype identity. Several transcription factors that are expressed mainly in precursors of the ventral telencephalon have been shown to control neuronal specification, but it has been unclear whether subtype identity is also specified in these precursors, or if this happens in postmitotic neurons, and whether it involves the same or different factors. SOX1, an HMG box transcription factor, is expressed widely in neural precursors along with the two other SOXB1 subfamily members, SOX2 and SOX3, and all three have been implicated in neurogenesis. SOX1 is also uniquely expressed at a high level in the majority of telencephalic neurons that constitute the ventral striatum (VS). These neurons are missing in Sox1-null mutant mice. In the present study, we have addressed the requirement for SOX1 at a cellular level, revealing both the nature and timing of the defect. By generating a novel Sox1-null allele expressing beta-galactosidase, we found that the VS precursors and their early neuronal differentiation are unaffected in the absence of SOX1, but the prospective neurons fail to migrate to their appropriate position. Furthermore, the migration of non-Sox1-expressing VS neurons (such as those expressing Pax6) was also affected in the absence of SOX1, suggesting that Sox1-expressing neurons play a role in structuring the area of the VS. To test whether SOX1 is required in postmitotic cells for the emergence of VS neuronal identity, we generated mice in which Sox1 expression was directed to all ventral telencephalic precursors, but to only a very few VS neurons. These mice again lacked most of the VS, indicating that SOX1 expression in precursors is not sufficient for VS development. Conversely, the few neurons in which Sox1 expression was maintained were able to migrate to the VS. In conclusion, Sox1 expression in precursors is not sufficient for VS neuronal identity and migration, but this is accomplished in postmitotic cells, which require the continued presence of SOX1. Our data also suggest that other SOXB1 members showing expression in specific neuronal populations are likely to play continuous roles from the establishment of precursors to their final differentiation.


Assuntos
Corpo Estriado/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas de Grupo de Alta Mobilidade/fisiologia , Neurônios/fisiologia , Telencéfalo/fisiologia , Animais , Movimento Celular/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Deleção de Genes , Vetores Genéticos , Proteínas de Grupo de Alta Mobilidade/deficiência , Proteínas de Grupo de Alta Mobilidade/genética , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Mapeamento por Restrição , Fatores de Transcrição SOXB1 , Proteína da Região Y Determinante do Sexo/genética
15.
Stem Cell Reports ; 8(3): 685-700, 2017 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28196689

RESUMO

Two populations of oligodendrogenic progenitors co-exist within the corpus callosum (CC) of the adult mouse. Local, parenchymal oligodendrocyte progenitor cells (pOPCs) and progenitors generated in the subependymal zone (SEZ) cytogenic niche. pOPCs are committed perinatally and retain their numbers through self-renewing divisions, while SEZ-derived cells are relatively "young," being constantly born from neural stem cells. We compared the behavior of these populations, labeling SEZ-derived cells using hGFAP:CreErt2 mice, within the homeostatic and regenerating CC of the young-adult and aging brain. We found that SEZ-derived oligodendroglial progenitors have limited self-renewing potential and are therefore not bona fide OPCs but rather "oligodendroblasts" more similar to the neuroblasts of the neurogenic output of the SEZ. In the aged CC their mitotic activity is much reduced, although they still act as a "fast-response element" to focal demyelination. In contrast to pOPCs, they fail to generate mature myelinating oligodendrocytes at all ages studied.


Assuntos
Doenças Desmielinizantes/etiologia , Doenças Desmielinizantes/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Fatores Etários , Animais , Biomarcadores , Encéfalo/citologia , Encéfalo/metabolismo , Diferenciação Celular , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Neurogênese , Nicho de Células-Tronco
16.
Front Cell Neurosci ; 10: 35, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26909025

RESUMO

Augmenting evidence suggests that such is the functional dependance of neural stem cells (NSCs) on the vasculature that they normally reside in "perivascular niches". Two examples are the "neurovascular" and the "oligovascular" niches of the adult brain, which comprise specialized microenvironments where NSCs or oligodendrocyte progenitor cells survive and remain mitotically active in close proximity to blood vessels (BVs). The often observed co-ordination of angiogenesis and neurogenesis led to these processes being described as "coupled". Here, we adopt an evo-devo approach to argue that some stages in the life of a NSC, such as specification and commitment, are independent of the vasculature, while stages such as proliferation and migration are largely dependent on BVs. We also explore available evidence on the possible involvement of the vasculature in other phenomena such as the diversification of NSCs during evolution and we provide original data on the senescence of NSCs in the subependymal zone stem cell niche. Finally, we will comment on the other side of the story; that is, on how much the vasculature is dependent on NSCs and their progeny.

17.
Front Biosci (Schol Ed) ; 8(1): 29-43, 2016 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26709894

RESUMO

Oligodendrocyte Progenitor Cells (OPCs) first appear at mid embryogenic stages during development of the mammalian CNS and a mitotically active population of them remains present even into late adulthood. During the life-time of the organism they initially proliferate and migrate in order to populate the whole nervous tissue, then they massively generate oligodendrocytesand finally they switch to a less mitotically active phase generating new oligodendrocytes at a slow rate in the adult brain; importantly, they can regenerate acutely or chronically destroyed myelin. All the above depend on the capacity of OPCs to regulate their cell cycle within different contexts. In this review we describe the development of OPCs, their differential mitotic behavior in various conditions (embryo, disease, ageing), we discuss what is known about the mechanisms that control their cell cycle and wehighlightfew interesting and still open questions.


Assuntos
Ciclo Celular , Sistema Nervoso Central/citologia , Oligodendroglia , Células-Tronco/fisiologia , Envelhecimento/fisiologia , Animais , Encéfalo , Diferenciação Celular , Humanos , Bainha de Mielina
18.
Exp Neurol ; 269: 75-89, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25819103

RESUMO

The presence of neural stem/progenitor cells (NSPCs) in specific areas of the central nervous system (CNS) supports tissue maintenance as well as regeneration. The subependymal zone (SEZ), located at the lateral ventricle's wall, represents a niche for NSPCs and in response to stroke or demyelination becomes activated with progenitors migrating towards the lesion and differentiating into neurons and glia. The mechanisms that underlie this phenomenon remain largely unknown. The vascular niche and in particular blood-derived elements such as platelets, has been shown to contribute to CNS regeneration in different pathological conditions. Indeed, intracerebroventricularly administrated platelet lysate (PL) stimulates angiogenesis, neurogenesis and neuroprotection in the damaged CNS. Here, we explored the presence of platelets in the activated SEZ after a focal demyelinating lesion in the corpus callosum of mice and we studied the effects of PL on proliferating SEZ-derived NSPCs in vitro. We showed that the lesion-induced increase in the size of the SEZ and in the number of proliferating SEZ-resident NSPCs correlates with the accumulation of platelets specifically along the activated SEZ vasculature. Expanding on this finding, we demonstrated that exposure of NSPCs to PL in vitro led to increased numbers of cells by enhanced cell survival and reduced apoptosis without differences in proliferation and in the differentiation potential of NSPCs. Finally, we demonstrate that the accumulation of platelets within the SEZ is spatially correlated with reduced numbers of apoptotic cells when compared to other periventricular areas. In conclusion, our results show that platelet-derived compounds specifically promote SEZ-derived NSPC survival and suggest that platelets might contribute to the enlargement of the pool of SEZ NSPCs that are available for CNS repair in response to injury.


Assuntos
Plaquetas/citologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Células-Tronco Adultas/citologia , Animais , Lesões Encefálicas/patologia , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Feminino , Masculino , Camundongos Endogâmicos C57BL , Neurônios/citologia
19.
Brain Res ; 991(1-2): 34-45, 2003 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-14575874

RESUMO

The elucidation of the molecular mechanisms involved in the response of brain tissue to trauma and the recognition of substances with neuroprotective properties is a prerequisite for the development of rational therapeutic approaches. In this study, we used a model of, unilateral, penetrating stab-like brain injury and examined the possible beneficial effects of post-injury administration of insulin-like growth factor-I (IGF-I) both at the cellular level, 4 and 12 h post-injury, and on the physical condition of the animals up to 1 week following the trauma. The consequences of injury were assessed by immunohistochemically observing the expression of heat-shock protein 70 (Hsp70), which is thought to be a marker of cell stress and injury, and by staining the tissue with the TUNEL reaction, in order to detect apoptotic cell death. Injury resulted in an increase in the number of Hsp70 and TUNEL positive cells in the peritraumatic area. The physical condition of the rats was followed by measuring body weight changes, food and water intake and by estimating their "motor activity". IGF-I administration resulted in a significant decrease in the number of Hsp70 and TUNEL positive cells in the peritraumatic area. Additionally, it improved the total "motor activity" of injured rats, increased food intake and attenuated the post-injury body weight loss. IGF-I thus emerges as a factor acting both at the cellular level as a neuroprotectant and at the systemic level as an anabolic agent.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/efeitos dos fármacos , Traumatismos Cranianos Penetrantes/tratamento farmacológico , Fator de Crescimento Insulin-Like I/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Choque Térmico HSP70/biossíntese , Traumatismos Cranianos Penetrantes/patologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Modelos Animais , Atividade Motora/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
20.
PLoS One ; 9(10): e108088, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25272038

RESUMO

Disrupted in schizophrenia 1 (DISC1) is a risk factor for a spectrum of neuropsychiatric illnesses including schizophrenia, bipolar disorder, and major depressive disorder. Here we use two missense Disc1 mouse mutants, described previously with distinct behavioural phenotypes, to demonstrate that Disc1 variation exerts differing effects on the formation of newly generated neurons in the adult hippocampus. Disc1 mice carrying a homozygous Q31L mutation, and displaying depressive-like phenotypes, have fewer proliferating cells while Disc1 mice with a homozygous L100P mutation that induces schizophrenia-like phenotypes, show changes in the generation, placement and maturation of newly generated neurons in the hippocampal dentate gyrus. Our results demonstrate Disc1 allele specific effects in the adult hippocampus, and suggest that the divergence in behavioural phenotypes may in part stem from changes in specific cell populations in the brain.


Assuntos
Variação Genética , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Animais , Movimento Celular/genética , Modelos Animais de Doenças , Expressão Gênica , Masculino , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Células-Tronco Neurais/metabolismo , Fenótipo , Esquizofrenia/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA