Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Development ; 148(3)2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33462112

RESUMO

Microtubules (MTs) regulate numerous cellular processes, but their roles in brain morphogenesis are not well known. Here, we show that CAMSAP3, a non-centrosomal microtubule regulator, is important for shaping the lateral ventricles. In differentiating ependymal cells, CAMSAP3 became concentrated at the apical domains, serving to generate MT networks at these sites. Camsap3-mutated mice showed abnormally narrow lateral ventricles, in which excessive stenosis or fusion was induced, leading to a decrease of neural stem cells at the ventricular and subventricular zones. This defect was ascribed at least in part to a failure of neocortical ependymal cells to broaden their apical domain, a process necessary for expanding the ventricular cavities. mTORC1 was required for ependymal cell growth but its activity was downregulated in mutant cells. Lysosomes, which mediate mTORC1 activation, tended to be reduced at the apical regions of the mutant cells, along with disorganized apical MT networks at the corresponding sites. These findings suggest that CAMSAP3 supports mTORC1 signaling required for ependymal cell growth via MT network regulation, and, in turn, shaping of the lateral ventricles.


Assuntos
Encéfalo/metabolismo , Ciclo Celular , Epêndima/crescimento & desenvolvimento , Ventrículos Laterais/crescimento & desenvolvimento , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Epêndima/metabolismo , Células Epiteliais/citologia , Feminino , Lisossomos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/metabolismo , Neuroglia/metabolismo
2.
Semin Cell Dev Biol ; 112: 61-68, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32771376

RESUMO

Within the adult mammalian central nervous system, the ventricular-subventricular zone (V-SVZ) lining the lateral ventricles houses neural stem cells (NSCs) that continue to produce neurons throughout life. Developmentally, the V-SVZ neurogenic niche arises during corticogenesis following the terminal differentiation of telencephalic radial glial cells (RGCs) into either adult neural stem cells (aNSCs) or ependymal cells. In mice, these two cellular populations form rosettes during the late embryonic and early postnatal period, with ependymal cells surrounding aNSCs. These aNSCs and ependymal cells serve a number of key purposes, including the generation of neurons throughout life (aNSCs), and acting as a barrier between the CSF and the parenchyma and promoting CSF bulk flow (ependymal cells). Interestingly, the development of this neurogenic niche, as well as its ongoing function, has been shown to be reliant on different aspects of lipid biology. In this review we discuss the developmental origins of the rodent V-SVZ neurogenic niche, and highlight research which has implicated a role for lipids in the physiology of this part of the brain. We also discuss the role of lipids in the maintenance of the V-SVZ niche, and discuss new research which has suggested that alterations to lipid biology could contribute to ependymal cell dysfunction in aging and disease.


Assuntos
Envelhecimento/genética , Epêndima/metabolismo , Lipídeos/genética , Células-Tronco Neurais/metabolismo , Envelhecimento/patologia , Animais , Proliferação de Células/genética , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Epêndima/crescimento & desenvolvimento , Epêndima/patologia , Humanos , Ventrículos Laterais/crescimento & desenvolvimento , Ventrículos Laterais/metabolismo , Ventrículos Laterais/patologia , Camundongos , Células-Tronco Neurais/fisiologia , Neurogênese/genética , Neurônios/metabolismo , Neurônios/patologia , Telencéfalo/metabolismo , Telencéfalo/patologia
3.
Fluids Barriers CNS ; 17(1): 72, 2020 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-33308296

RESUMO

BACKGROUND: Hydrocephalus, the pathological expansion of the cerebrospinal fluid (CSF)-filled cerebral ventricles, is a common, deadly disease. In the adult, cardiac and respiratory forces are the main drivers of CSF flow within the brain ventricular system to remove waste and deliver nutrients. In contrast, the mechanics and functions of CSF circulation in the embryonic brain are poorly understood. This is primarily due to the lack of model systems and imaging technology to study these early time points. Here, we studied embryos of the vertebrate Xenopus with optical coherence tomography (OCT) imaging to investigate in vivo ventricular and neural development during the onset of CSF circulation. METHODS: Optical coherence tomography (OCT), a cross-sectional imaging modality, was used to study developing Xenopus tadpole brains and to dynamically detect in vivo ventricular morphology and CSF circulation in real-time, at micrometer resolution. The effects of immobilizing cilia and cardiac ablation were investigated. RESULTS: In Xenopus, using OCT imaging, we demonstrated that ventriculogenesis can be tracked throughout development until the beginning of metamorphosis. We found that during Xenopus embryogenesis, initially, CSF fills the primitive ventricular space and remains static, followed by the initiation of the cilia driven CSF circulation where ependymal cilia create a polarized CSF flow. No pulsatile flow was detected throughout these tailbud and early tadpole stages. As development progressed, despite the emergence of the choroid plexus in Xenopus, cardiac forces did not contribute to the CSF circulation, and ciliary flow remained the driver of the intercompartmental bidirectional flow as well as the near-wall flow. We finally showed that cilia driven flow is crucial for proper rostral development and regulated the spatial neural cell organization. CONCLUSIONS: Our data support a paradigm in which Xenopus embryonic ventriculogenesis and rostral brain development are critically dependent on ependymal cilia-driven CSF flow currents that are generated independently of cardiac pulsatile forces. Our work suggests that the Xenopus ventricular system forms a complex cilia-driven CSF flow network which regulates neural cell organization. This work will redirect efforts to understand the molecular regulators of embryonic CSF flow by focusing attention on motile cilia rather than other forces relevant only to the adult.


Assuntos
Encéfalo/crescimento & desenvolvimento , Líquido Cefalorraquidiano/fisiologia , Cílios , Epêndima/crescimento & desenvolvimento , Coração/fisiologia , Larva/crescimento & desenvolvimento , Xenopus/crescimento & desenvolvimento , Animais , Fluxo Pulsátil/fisiologia , Tomografia de Coerência Óptica
4.
Sci Rep ; 10(1): 12435, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32709945

RESUMO

Hydrocephalus is a brain disorder triggered by cerebrospinal fluid accumulation in brain cavities. Even though cerebrospinal fluid flow is known to be driven by the orchestrated beating of the bundled motile cilia of ependymal cells, little is known about the mechanism of ciliary motility. RSPH9 is increasingly becoming recognized as a vital component of radial spokes in ciliary "9 + 2" ultrastructure organization. Here, we show that deletion of the Rsph9 gene leads to the development of hydrocephalus in the early postnatal period. However, the neurodevelopment and astrocyte development are normal in embryonic Rsph9-/- mice. The tubular structure of the central aqueduct was comparable in Rsph9-/- mice. Using high-speed video microscopy, we visualized lower beating amplitude and irregular rotation beating pattern of cilia bundles in Rsph9-/- mice compared with that of wild-type mice. And the centriolar patch size was significantly increased in Rsph9-/- cells. TEM results showed that deletion of Rsph9 causes little impact in ciliary axonemal organization but the Rsph9-/- cilia frequently had abnormal ectopic ciliary membrane inclusions. In addition, hydrocephalus in Rsph9-/- mice results in the development of astrogliosis, microgliosis and cerebrovascular abnormalities. Eventually, the ependymal cells sloughed off of the lateral wall. Our results collectively suggested that RSPH9 is essential for ciliary structure and motility of mouse ependymal cilia, and its deletion causes the pathogenesis of hydrocephalus.


Assuntos
Cílios/patologia , Proteínas do Citoesqueleto/genética , Epêndima/crescimento & desenvolvimento , Hidrocefalia/genética , Animais , Animais Recém-Nascidos , Axonema/ultraestrutura , Cílios/metabolismo , Cílios/ultraestrutura , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Epêndima/citologia , Epêndima/patologia , Epêndima/ultraestrutura , Feminino , Humanos , Hidrocefalia/congênito , Hidrocefalia/patologia , Microscopia Intravital , Masculino , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Microscopia de Vídeo
5.
Sci Rep ; 10(1): 2826, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32071335

RESUMO

Tanycyte is a subtype of ependymal cells which extend long radial processes to brain parenchyma. The present study showed that tanycyte-like ependymal cells in the organum vasculosum of the lamina terminalis, subfornical organ and central canal (CC) expressed neural stem cell (NSC) marker nestin, glial fibrillar acidic protein and sex determining region Y. Proliferation of these tanycyte-like ependymal cells was promoted by continuous intracerebroventricular infusion of fibroblast growth factor-2 and epidermal growth factor. Tanycytes-like ependymal cells in the CC are able to form self-renewing neurospheres and give rise mostly to new astrocytes and oligodendrocytes. Collagenase-induced small medullary hemorrhage increased proliferation of tanycyte-like ependymal cells in the CC. These results demonstrate that these tanycyte-like ependymal cells of the adult mouse brain are NSCs and suggest that they serve as a source for providing new neuronal lineage cells upon brain damage in the medulla oblongata.


Assuntos
Órgãos Circunventriculares/metabolismo , Células Ependimogliais/metabolismo , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Linhagem da Célula/genética , Proliferação de Células/genética , Órgãos Circunventriculares/crescimento & desenvolvimento , Epêndima/crescimento & desenvolvimento , Epêndima/metabolismo , Células Ependimogliais/citologia , Fator de Crescimento Epidérmico/genética , Fator 2 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica/genética , Humanos , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Camundongos , Nestina/genética , Células-Tronco Neurais/citologia , Organum Vasculosum/crescimento & desenvolvimento , Organum Vasculosum/metabolismo , Órgão Subfornical/crescimento & desenvolvimento , Órgão Subfornical/metabolismo
6.
J Neurosci ; 40(11): 2246-2258, 2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-32001613

RESUMO

The ependyma of the adult spinal cord is a latent stem cell niche that is reactivated by spinal cord injury contributing new cells to the glial scar. The cellular events taking place in the early stages of the reaction of the ependyma to injury remain little understood. Ependymal cells are functionally heterogeneous with a mitotically active subpopulation lining the lateral domains of the central canal (CC) that are coupled via gap junctions. Gap junctions and connexin hemichannels are key regulators of the biology of neural progenitors during development and in adult neurogenic niches. Thus, we hypothesized that communication via connexins in the CC is developmentally regulated and may play a part in the reactivation of this latent stem cell niche after injury. To test these possibilities, we combined patch-clamp recordings of ependymal cells with immunohistochemistry for various connexins in the neonatal and the adult (P > 90) normal and injured spinal cord of male and female mice. We find that coupling among ependymal cells is downregulated as postnatal development proceeds but increases after injury, resembling the immature CC. The increase in gap junction coupling in the adult CC was paralleled by upregulation of connexin 26, which correlated with the resumption of proliferation and a reduction of connexin hemichannel activity. Connexin blockade reduced the injury-induced proliferation of ependymal cells. Our findings suggest that connexins are involved in the early reaction of ependymal cells to injury, representing a potential target to improve the contribution of the CC stem cell niche to repair.SIGNIFICANCE STATEMENT Ependymal cells in the adult spinal cord are latent progenitors that react to injury to support some degree of endogenous repair. Understanding the mechanisms by which these progenitor-like cells are regulated in the aftermath of spinal cord injury is critical to design future manipulations aimed at improving healing and functional recovery. Gap junctions and connexin hemichannels are key regulators of the biology of neural progenitors during development and in adult neurogenic niches. We find here that connexin signaling in the ependyma changes after injury of the adult spinal cord, functionally resembling the immature active-stem cell niche of neonatal animals. Our findings suggest that connexins in ependymal cells are potential targets to improve self-repair of the spinal cord.


Assuntos
Conexinas/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Traumatismos da Medula Espinal/fisiopatologia , Nicho de Células-Tronco/fisiologia , Fatores Etários , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Membrana Celular/fisiologia , Permeabilidade da Membrana Celular , Conexinas/antagonistas & inibidores , Epêndima/citologia , Epêndima/crescimento & desenvolvimento , Feminino , Corantes Fluorescentes/farmacocinética , Junções Comunicantes/fisiologia , Hidrogéis , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/antagonistas & inibidores , Técnicas de Patch-Clamp , Peptídeos/química , Peptídeos/farmacologia , Poloxâmero/farmacologia , Distribuição Aleatória
7.
Mol Cells ; 42(3): 245-251, 2019 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-30759972

RESUMO

Ependymal cells constitute the multi-ciliated epithelium, which lines the brain ventricular lumen. Although ependymal cells originate from radial glial cells in the perinatal rodent brain, the exact mechanisms underlying the full differentiation of ependymal cells are poorly understood. In this report, we present evidence that the Anks1a phosphotyrosine binding domain (PTB) adaptor is required for the proper development of ependymal cells in the rodent postnatal brain. Anks1a gene trap targeted LacZ reporter analysis revealed that Anks1a is expressed prominently in the ventricular region of the early postnatal brain and that its expression is restricted to mature ependymal cells during postnatal brain development. In addition, Anks1a-deficient ependymal cells were shown to possess type B cell characteristics, suggesting that ependymal cells require Anks1a in order to be fully differentiated. Finally, Anks1a overexpression in the lateral wall of the neonatal brain resulted in an increase in the number of ependymal cells during postnatal brain development. Altogether, our results suggest that ependymal cells require Anks1a PTB adaptor for their proper development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Epêndima/citologia , Epêndima/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Contagem de Células , Diferenciação Celular , Cromossomos Artificiais Bacterianos/genética , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos Transgênicos , Regulação para Cima
8.
Nat Commun ; 9(1): 2279, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29891944

RESUMO

Multiciliated ependymal cells line all brain cavities. The beating of their motile cilia contributes to the flow of cerebrospinal fluid, which is required for brain homoeostasis and functions. Motile cilia, nucleated from centrioles, persist once formed and withstand the forces produced by the external fluid flow and by their own cilia beating. Here, we show that a dense actin network around the centrioles is induced by cilia beating, as shown by the disorganisation of the actin network upon impairment of cilia motility. Moreover, disruption of the actin network, or specifically of the apical actin network, causes motile cilia and their centrioles to detach from the apical surface of ependymal cell. In conclusion, cilia beating controls the apical actin network around centrioles; the mechanical resistance of this actin network contributes, in turn, to centriole stability.


Assuntos
Actinas/fisiologia , Centríolos/fisiologia , Cílios/fisiologia , Epêndima/fisiologia , Actinas/química , Animais , Fenômenos Biomecânicos , Proteínas do Citoesqueleto , Epêndima/crescimento & desenvolvimento , Epêndima/ultraestrutura , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas dos Microfilamentos , Modelos Neurológicos , Mapas de Interação de Proteínas , Proteínas/genética , Proteínas/metabolismo
9.
J Neurosci ; 38(14): 3466-3479, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29507144

RESUMO

Exposure of the developing brain to toxins, drugs, or deleterious endogenous compounds during the perinatal period can trigger alterations in cell division, migration, differentiation, and synaptogenesis, leading to lifelong neurological impairment. The brain is protected by cellular barriers acting through multiple mechanisms, some of which are still poorly explored. We used a combination of enzymatic assays, live tissue fluorescence microscopy, and an in vitro cellular model of the blood-CSF barrier to investigate an enzymatic detoxification pathway in the developing male and female rat brain. We show that during the early postnatal period the choroid plexus epithelium forming the blood-CSF barrier and the ependymal cell layer bordering the ventricles harbor a high detoxifying capacity that involves glutathione S-transferases. Using a functional knock-down rat model for choroidal glutathione conjugation, we demonstrate that already in neonates, this metabolic pathway efficiently prevents the penetration of blood-borne reactive compounds into CSF. The versatility of the protective mechanism results from the multiplicity of the glutathione S-transferase isoenzymes, which are differently expressed between the choroidal epithelium and the ependyma. The various isoenzymes display differential substrate specificities, which greatly widen the spectrum of molecules that can be inactivated by this pathway. In conclusion, the blood-CSF barrier and the ependyma are identified as key cellular structures in the CNS to protect the brain fluid environment from different chemical classes of potentially toxic compounds during the postnatal period. This metabolic neuroprotective function of brain interfaces ought to compensate for the liver postnatal immaturity.SIGNIFICANCE STATEMENT Brain homeostasis requires a stable and controlled internal environment. Defective brain protection during the perinatal period can lead to lifelong neurological impairment. We demonstrate that the choroid plexus forming the blood-CSF barrier is a key player in the protection of the developing brain. Glutathione-dependent enzymatic metabolism in the choroidal epithelium inactivates a broad spectrum of noxious compounds, efficiently preventing their penetration into the CSF. A second line of detoxification is located in the ependyma separating the CSF from brain tissue. Our study reveals a novel facet of the mechanisms by which the brain is protected at a period of high vulnerability, at a time when the astrocytic network is still immature and liver xenobiotic metabolism is limited.


Assuntos
Barreira Hematoencefálica/metabolismo , Glutationa Transferase/metabolismo , Glutationa/metabolismo , Animais , Barreira Hematoencefálica/crescimento & desenvolvimento , Plexo Corióideo/crescimento & desenvolvimento , Plexo Corióideo/metabolismo , Epêndima/crescimento & desenvolvimento , Epêndima/metabolismo , Feminino , Radicais Livres/sangue , Radicais Livres/líquido cefalorraquidiano , Glutationa/sangue , Glutationa/líquido cefalorraquidiano , Masculino , Ratos , Ratos Sprague-Dawley
10.
J Comp Neurol ; 526(8): 1267-1286, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29405286

RESUMO

Nervous system development is a precisely orchestrated series of events requiring a multitude of intrinsic and extrinsic cues. Sortilin and SorCS2 are members of the Vps10p receptor family with complementary influence on some of these cues including the neurotrophins (NTs). However, the developmental time points where sortilin and SorCS2 exert their activities in conjunction or independently still remain unclear. In this study we present the characterization of the spatiotemporal expression pattern of sortilin and SorCS2 in the developing murine nervous system. Sortilin is highly expressed in the fetal nervous system with expression localized to distinct cell populations. Expression was high in neurons of the cortical plate and developing allocortex, as well as subpallial structures. Furthermore, the neuroepithelium lining the ventricles and the choroid plexus showed high expression of sortilin, together with the developing retina, spinal ganglia, and sympathetic ganglia. In contrast, SorCS2 was confined in a marked degree to the thalamus and, at E13.5, the floor plate from midbrain rostrally to spinal cord caudally. SorCS2 was also found in the ventricular zones of the ventral hippocampus and nucleus accumbens areas, in the meninges and in Schwann cells. Hence, sortilin and SorCS2 are extensively present in several distinct anatomical areas in the developing nervous system and are rarely co-expressed. Possible functions of sortilin and SorCS2 pertain to NT signaling, axon guidance and beyond. The present data will form the basis for hypotheses and study designs for unravelling the functions of sortilin and SorCS2 during the establishment of neuronal structures and connections.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Sistema Nervoso Central , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Nervos Periféricos , Receptores de Superfície Celular/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Animais Recém-Nascidos , Calbindina 2/metabolismo , Calbindinas/metabolismo , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Plexo Corióideo/embriologia , Plexo Corióideo/crescimento & desenvolvimento , Plexo Corióideo/metabolismo , Embrião de Mamíferos , Epêndima/embriologia , Epêndima/crescimento & desenvolvimento , Epêndima/metabolismo , Meninges/embriologia , Meninges/crescimento & desenvolvimento , Meninges/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Nervos Periféricos/embriologia , Nervos Periféricos/crescimento & desenvolvimento , Nervos Periféricos/metabolismo , Receptores de Superfície Celular/genética , Tirosina 3-Mono-Oxigenase/metabolismo
11.
J Comp Neurol ; 525(3): 693-707, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27503700

RESUMO

According to previous opinion, the derivation of neurons and glia from the central canal (CC) lining of the spinal cord in rodents should occur in the embryonic period. Reports of the mitotic activity observed in the lining during postnatal development have often been contradictory, and proliferation was ascribed to the generation of ependymocytes, which are necessary for the elongation of CC walls. Our study quantifies the intensity of proliferation and determines the cellularity of the CC lining in reference to lumbar spinal segment L4 during the postnatal development of rats. The presence of dividing cells peaks in the CC lining on postnatal day 8 (P8), with division occurring in 19.2% ± 3.2% of cells. In adult rats, 3.6% ± 0.9% of cells still proliferate, whereas, in mice, 10.3% ± 2.3% of cells at P8 and only 0.6% ± 0.2% of cells in the CC lining in adulthood are proliferating. In the rat, the length of the cell cycle increases from 100.3 ± 35.7 hours at P1 to 401.4 ± 80.6 hours at P43, with a sudden extension between P15 and P22. Despite the intensive proliferation, the total cellularity of the CC lining at the L4 spinal segment significantly descended in from P8 to P15. According to our calculations, the estimated cellularity was significantly higher compared with the measured cellularity of the CC lining at P15. Our results indicate that CC lining serves as a source of cells beyond ependymal cells during the first postnatal weeks of the rat. J. Comp. Neurol. 525:693-707, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Proliferação de Células , Medula Espinal/citologia , Medula Espinal/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Bromodesoxiuridina , Ciclo Celular , Epêndima/citologia , Epêndima/crescimento & desenvolvimento , Imunofluorescência , Antígeno Ki-67/metabolismo , Vértebras Lombares , Camundongos Endogâmicos BALB C , Microscopia Confocal , Neuroglia/citologia , Neurônios/citologia , Ratos Wistar , Especificidade da Espécie , Fatores de Tempo
12.
Trends Neurosci ; 39(8): 543-551, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27311928

RESUMO

Cerebrospinal fluid (CSF) continuously flows through the cerebral ventricles, a process essential for brain homeostasis. Multiciliated ependymal (E1) cells line the walls of the ventricles and contribute importantly to CSF flow through ciliary beating. Key to this function is the rotational and translational planar cell polarity (PCP) of E1 cells. Defects in the PCP of E1 cells can result in abnormal CSF accumulation and hydrocephalus. Here, we integrate recent data on the roles of early CSF flow in the embryonic ventricles, PCP regulators (e.g., Vangl2 and Dishevelled), and cytoskeletal networks in the establishment, refinement, and maintenance of E1 cells' PCP. The planar organization mechanisms of E1 cells could explain how CSF flow contributes to brain function and may help in the diagnosis and prevention of hydrocephalus.


Assuntos
Epêndima/citologia , Epêndima/fisiologia , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Animais , Epêndima/crescimento & desenvolvimento , Epêndima/fisiopatologia , Humanos , Hidrocefalia/patologia , Hidrocefalia/fisiopatologia
13.
Differentiation ; 91(4-5): 28-41, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27016251

RESUMO

Individual cells dissected from the subependymal neurogenic niche of the adult mouse brain proliferate in medium containing basic fibroblast growth factor (bFGF) and/or epidermal growth factor (EGF) as mitogens, to produce multipotent clonal aggregates called neurospheres. These cultures constitute a powerful tool for the study of neural stem cells (NSCs) provided that they allow the analysis of their features and potential capacity in a controlled environment that can be modulated and monitored more accurately than in vivo. Clonogenic and population analyses under mitogen addition or withdrawal allow the quantification of the self-renewing and multilineage potency of these cells and the identification of the mechanisms involved in these properties. Here, we describe a set of procedures developed and/or modified by our group including several experimental options that can be used either independently or in combination for the ex vivo assessment of cell properties of NSCs obtained from the adult subependymal niche.


Assuntos
Técnicas de Cultura de Células , Epêndima/crescimento & desenvolvimento , Células-Tronco Neurais/citologia , Neurogênese/genética , Células-Tronco Adultas , Animais , Diferenciação Celular/genética , Epêndima/citologia , Humanos , Camundongos , Neurônios
14.
J Comp Neurol ; 524(15): 2982-92, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-26991819

RESUMO

In mammals, ventricular walls of the developing brain maintain a neurogenic niche, in which radial glial cells act as neural stem cells (NSCs) and generate new neurons in the embryo. In the adult brain, the neurogenic niche is maintained in the ventricular-subventricular zone (V-SVZ) of the lateral wall of lateral ventricles and the hippocampal dentate gyrus. In the neonatal V-SVZ, radial glial cells transform into astrocytic postnatal NSCs and multiciliated ependymal cells. On the other hand, in zebrafish, radial glial cells continue to cover the surface of the adult telencephalic ventricle and maintain a higher neurogenic potential in the adult brain. However, the cell composition of the neurogenic niche of the aged zebrafish brain has not been investigated. Here we show that multiciliated ependymal cells emerge in the neurogenic niche of the aged zebrafish telencephalon. These multiciliated cells appear predominantly in the dorsal part of the ventral telencephalic ventricular zone, which also contains clusters of migrating new neurons. Scanning electron microscopy and live imaging analyses indicated that these multiple cilia beat coordinately and generate constant fluid flow within the ventral telencephalic ventricle. Analysis of the cell composition by transmission electron microscopy revealed that the neurogenic niche in the aged zebrafish contains different types of cells, with ultrastructures similar to those of ependymal cells, transit-amplifying cells, and migrating new neurons in postnatal mice. These data suggest that the transformation capacity of radial glial cells is conserved but that its timing is different between fish and mice. J. Comp. Neurol. 524:2982-2992, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Envelhecimento/fisiologia , Epêndima/citologia , Nicho de Células-Tronco/fisiologia , Telencéfalo/citologia , Peixe-Zebra/fisiologia , Envelhecimento/patologia , Animais , Animais Geneticamente Modificados , Movimento Celular/fisiologia , Cílios/ultraestrutura , Epêndima/crescimento & desenvolvimento , Epêndima/fisiologia , Epêndima/ultraestrutura , Imuno-Histoquímica , Microscopia Confocal , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Telencéfalo/crescimento & desenvolvimento , Telencéfalo/fisiologia , Telencéfalo/ultraestrutura , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/crescimento & desenvolvimento
15.
Brain Res ; 1616: 71-87, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25960350

RESUMO

Nuclear factor one X (NFIX) has been shown to play a pivotal role during the development of many regions of the brain, including the neocortex, the hippocampus and the cerebellum. Mechanistically, NFIX has been shown to promote neural stem cell differentiation through the activation of astrocyte-specific genes and via the repression of genes central to progenitor cell self-renewal. Interestingly, mice lacking Nfix also exhibit other phenotypes with respect to development of the central nervous system, and whose underlying causes have yet to be determined. Here we examine one of the phenotypes displayed by Nfix(-/-) mice, namely hydrocephalus. Through the examination of embryonic and postnatal Nfix(-/-) mice we reveal that hydrocephalus is first seen at around postnatal day (P) 10 in mice lacking Nfix, and is fully penetrant by P20. Furthermore, we examined the subcommissural organ (SCO), the Sylvian aqueduct and the ependymal layer of the lateral ventricles, regions that when malformed and functionally perturbed have previously been implicated in the development of hydrocephalus. SOX3 is a factor known to regulate SCO development. Although we revealed that NFIX could repress Sox3-promoter-driven transcriptional activity in vitro, SOX3 expression within the SCO was normal within Nfix(-/-) mice, and Nfix mutant mice showed no abnormalities in the structure or function of the SCO. Moreover, these mutant mice exhibited no overt blockage of the Sylvian aqueduct. However, the ependymal layer of the lateral ventricles was frequently absent in Nfix(-/-) mice, suggesting that this phenotype may underlie the development of hydrocephalus within these knockout mice.


Assuntos
Epêndima/patologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Hidrocefalia/patologia , Ventrículos Laterais/patologia , Fatores de Transcrição NFI/deficiência , Fatores Etários , Animais , Animais Recém-Nascidos , Biologia Computacional , Modelos Animais de Doenças , Embrião de Mamíferos , Epêndima/embriologia , Epêndima/crescimento & desenvolvimento , Hidrocefalia/genética , Ventrículos Laterais/embriologia , Ventrículos Laterais/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores de Transcrição NFI/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo
16.
J Comp Neurol ; 522(11): 2663-79, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24500610

RESUMO

Trp73, a member of the p53 gene family, plays a crucial role in neural development. We describe two main phenotypic variants of p73 deficiency in the brain, a severe one characterized by massive apoptosis in the cortex leading to early postnatal death and a milder, non-/low-apoptosis one in which 50% of pups may reach adulthood using an intensive-care breeding protocol. Both variants display the core triad of p73 deficiency: cortical hypoplasia, hippocampal malformations, and ventriculomegaly. We studied the development of the neocortex in p73 KO mice from early embryonic life into advanced age (25 months). Already at E14.5, the incipient cortical plate of the p73 KO brains showed a reduced width. Examination of adult neocortex revealed a generalized, nonprogressive reduction by 10-20%. Area-specific architectonic landmarks and lamination were preserved in all cortical areas. The surviving adult animals had moderate ventricular distension, whereas pups of the early lethal phenotypic variant showed severe ventriculomegaly. Ependymal cells of wild-type ventricles strongly express p73 and are particularly vulnerable to p73 deficiency. Ependymal denudation by apoptosis and reduction of ependymal cilia were already evident in young mice, with complete absence of cilia in older animals. Loss of p73 function in the ependyma may thus be one determining factor for chronic hydrocephalus, which leads to atrophy of subcortical structures (striatum, septum, amygdala). p73 Is thus involved in a variety of CNS activities ranging from embryonic regulation of brain size to the control of cerebrospinal fluid homeostasis in the adult brain via maintenance of the ependyma.


Assuntos
Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/fisiologia , Hidrocefalia/fisiopatologia , Neocórtex/anormalidades , Neocórtex/crescimento & desenvolvimento , Proteínas Nucleares/deficiência , Proteínas Nucleares/fisiologia , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/fisiologia , Animais , Apoptose , Contagem de Células , Plexo Corióideo/anormalidades , Plexo Corióideo/crescimento & desenvolvimento , Plexo Corióideo/fisiopatologia , Proteínas de Ligação a DNA/genética , Epêndima/anormalidades , Epêndima/crescimento & desenvolvimento , Epêndima/fisiopatologia , Imunofluorescência , Hipocampo/anormalidades , Hipocampo/crescimento & desenvolvimento , Hipocampo/fisiopatologia , Hidrocefalia/patologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Neocórtex/fisiopatologia , Proteínas Nucleares/genética , Fenótipo , Proteína Tumoral p73 , Proteínas Supressoras de Tumor/genética
17.
Morfologiia ; 146(5): 70-2, 2014.
Artigo em Russo | MEDLINE | ID: mdl-25823294

RESUMO

It is known that during development of the brain, with the progress of ependendymocyte differentiation from radial gliocytes, the synthesis of nestin is stopped. However, it was shown that in the ependyma of the lateral brain ventricles nestin synthesis was resumed in response to ischemic injury. The aim of the present study was to test the hypothesis of possible re-expression of nestin in the ependyma in aging. The study was performed on male Wistar rats aged 4 (n = 4) and 28 months (n = 3). In older animals the expression of nestin was demonstrated in the ependyma of the lateral ventricles of the brain. It was also found that the area of the medial and upper walls of the lateral ventricle contained the regions of ependyma, in which all cells had intense cytoplasmic staining. The causes of the phenomenon described remain unclear.


Assuntos
Envelhecimento/metabolismo , Epêndima/metabolismo , Ventrículos Laterais/metabolismo , Nestina/metabolismo , Animais , Epêndima/citologia , Epêndima/crescimento & desenvolvimento , Ventrículos Laterais/citologia , Ventrículos Laterais/crescimento & desenvolvimento , Masculino , Nestina/genética , Ratos , Ratos Wistar
18.
Dev Biol ; 382(1): 172-85, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23906841

RESUMO

Congenital hydrocephalus, the accumulation of excess cerebrospinal fluid (CSF) in the ventricles of the brain, affects one of every 1000 children born today, making it one of the most common human developmental disorders. Genetic causes of hydrocephalus are poorly understood in humans, but animal models suggest a broad genetic program underlying the regulation of CSF balance. In this study, the random integration of a transgene into the mouse genome led to the development of an early onset and rapidly progressive hydrocephalus. Juvenile hydrocephalus transgenic mice (Jhy(lacZ)) inherit communicating hydrocephalus in an autosomal recessive fashion with dilation of the lateral ventricles observed as early as postnatal day 1.5. Ventricular dilation increases in severity over time, becoming fatal at 4-8 weeks of age. The ependymal cilia lining the lateral ventricles are morphologically abnormal and reduced in number in Jhy(lacZ/lacZ) brains, and ultrastructural analysis revealed disorganization of the expected 9+2 microtubule pattern. Rather, the majority of Jhy(lacZ/lacZ) cilia develop axonemes with 9+0 or 8+2 microtubule structures. Disruption of an unstudied gene, 4931429I11Rik (now named Jhy) appears to underlie the hydrocephalus of Jhy(lacZ/lacZ) mice, and the Jhy transcript and protein are decreased in Jhy(lacZ/lacZ) mice. Partial phenotypic rescue was achieved in Jhy(lacZ/lacZ) mice by the introduction of a bacterial artificial chromosome (BAC) carrying 60-70% of the JHY protein coding sequence. Jhy is evolutionarily conserved from humans to basal vertebrates, but the predicted JHY protein lacks identifiable functional domains. Ongoing studies are directed at uncovering the physiological function of JHY and its role in CSF homeostasis.


Assuntos
Cílios/patologia , Deleção de Genes , Hidrocefalia/genética , Hidrocefalia/patologia , Microtúbulos/metabolismo , Proteínas/genética , Envelhecimento , Sequência de Aminoácidos , Animais , Sequência de Bases , Líquido Cefalorraquidiano/metabolismo , Cílios/metabolismo , Cílios/ultraestrutura , Progressão da Doença , Elementos Facilitadores Genéticos/genética , Epêndima/crescimento & desenvolvimento , Epêndima/metabolismo , Epêndima/patologia , Epêndima/ultraestrutura , Genes Reporter , Humanos , Hidrocefalia/líquido cefalorraquidiano , Camundongos , Camundongos Transgênicos , Microtúbulos/ultraestrutura , Dados de Sequência Molecular , Fenótipo , Proteínas/química , Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Síndrome , Transgenes/genética
19.
Neurosci Lett ; 553: 57-62, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-23872091

RESUMO

The area surrounding the central canal of the postnatal mammalian spinal cord is a highly plastic region that exhibits many similarities to other postnatal neurogenic niches, such as the subventricular zone. Within this region, ependymal cells have been identified as neural stem cells however very little is known about their properties and how the local environment, including neurotransmitters, is capable of affecting them. The neurotransmitter GABA is present around the central canal and is known to affect cells within other postnatal neurogenic niches. This study used whole cell patch clamp electrophysiology and intracellular dye-loading in in vitro Wistar rat spinal cord slices to characterise ependymal cells and their ability to respond to GABA. Ependymal cells were defined by their passive response properties and low input resistances. Extensive dye-coupling was observed between ependymal cells; this was confirmed as gap junction coupling using the gap junction blocker, 18ß-glycyrrhetinic acid, which significantly increased the input resistance of ependymal cells. GABA depolarised all ependymal cells tested; the partial antagonism of this response by bicuculline and gabazine indicates that GABA(A) receptors contribute to this response. A lack of effect by baclofen suggests that GABA(B) receptors do not contribute to the GABAergic response. The ability of ependymal cells to respond to GABA suggests that GABA could be capable of influencing the proliferation and differentiation of cells within the neurogenic niche of the postnatal spinal cord.


Assuntos
Epêndima/metabolismo , Células-Tronco Neurais/fisiologia , Receptores de GABA-A/metabolismo , Medula Espinal/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Corantes , Epêndima/citologia , Epêndima/crescimento & desenvolvimento , Junções Comunicantes/fisiologia , Técnicas In Vitro , Técnicas de Patch-Clamp , Ratos Wistar , Medula Espinal/crescimento & desenvolvimento , Ácido gama-Aminobutírico/farmacologia
20.
Development ; 138(24): 5291-300, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22071110

RESUMO

Ependymal cells are part of the neurogenic niche in the adult subventricular zone of the lateral ventricles, where they regulate neurogenesis and neuroblast migration. Ependymal cells are generated from radial glia cells during embryonic brain development and acquire their final characteristics postnatally. The homeobox gene Six3 is expressed in ependymal cells during the formation of the lateral wall of the lateral ventricles in the brain. Here, we show that Six3 is necessary for ependymal cell maturation during postnatal stages of brain development. In its absence, ependymal cells fail to suppress radial glia characteristics, resulting in a defective lateral wall, abnormal neuroblast migration and differentiation, and hydrocephaly.


Assuntos
Epêndima/crescimento & desenvolvimento , Proteínas do Olho/fisiologia , Proteínas de Homeodomínio/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Animais , Encéfalo/crescimento & desenvolvimento , Diferenciação Celular , Movimento Celular , Proteínas do Olho/genética , Proteínas de Homeodomínio/genética , Hidrocefalia/etiologia , Ventrículos Laterais/crescimento & desenvolvimento , Camundongos , Camundongos Mutantes , Proteínas do Tecido Nervoso/genética , Neuroglia/fisiologia , Proteína Homeobox SIX3
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA