Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Development ; 137(18): 3037-46, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20685736

RESUMO

Motile cilia generate constant fluid flow over epithelial tissue, and thereby influence diverse physiological processes. Such functions of ciliated cells depend on the planar polarity of the cilia and on their basal bodies being oriented in the downstream direction of fluid flow. Recently, another type of basal body planar polarity, characterized by the anterior localization of the basal bodies in individual cells, was reported in the multiciliated ependymal cells that line the surface of brain ventricles. However, little is known about the cellular and molecular mechanisms by which this polarity is established. Here, we report in mice that basal bodies move in the apical cell membrane during differentiation to accumulate in the anterior region of ependymal cells. The planar cell polarity signaling pathway influences basal body orientation, but not their anterior migration, in the neonatal brain. Moreover, we show by pharmacological and genetic studies that non-muscle myosin II is a key regulator of this distribution of basal bodies. This study demonstrates that the orientation and distribution of basal bodies occur by distinct mechanisms.


Assuntos
Movimento Celular , Polaridade Celular , Epêndima/crescimento & desenvolvimento , Epêndima/metabolismo , Miosina Tipo II/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Cílios/metabolismo , Epêndima/citologia , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos ICR , Microscopia Eletrônica de Varredura , Miosina Tipo II/genética , Biossíntese de Proteínas
10.
Acta Neuropathol ; 121(6): 721-35, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21311902

RESUMO

A heterogeneous population of ependymal cells lines the brain ventricles. The evidence about the origin and birth dates of these cell populations is scarce. Furthermore, the possibility that mature ependymal cells are born (ependymogenesis) or self-renewed (ependymal proliferation) postnatally is controversial. The present study was designed to investigate both phenomena in wild-type (wt) and hydrocephalic α-SNAP mutant (hyh) mice at different postnatal stages. In wt mice, proliferating cells in the ventricular zone (VZ) were only found in two distinct regions: the dorsal walls of the third ventricle and Sylvian aqueduct (SA). Most proliferating cells were monociliated and nestin+, likely corresponding to radial glial cells. Postnatal cumulative BrdU-labeling showed that most daughter cells remained in the VZ of both regions and they lost nestin-immunoreactivity. Furthermore, some labeled cells became multiciliated and GLUT-1+, indicating they were ependymal cells born postnatally. Postnatal pulse BrdU-labeling and Ki-67 immunostaining further demonstrated the presence of cycling multiciliated ependymal cells. In hydrocephalic mutants, the dorsal walls of the third ventricle and SA expanded enormously and showed neither ependymal disruption nor ventriculostomies. This phenomenon was sustained by an increased ependymogenesis. Consequently, in addition to the physical and geometrical mechanisms traditionally explaining ventricular enlargement in fetal-onset hydrocephalus, we propose that postnatal ependymogenesis could also play a role. Furthermore, as generation of new ependymal cells during postnatal stages was observed in distinct regions of the ventricular walls, such as the roof of the third ventricle, it may be a key mechanism involved in the development of human type 1 interhemispheric cysts.


Assuntos
Encéfalo/patologia , Epêndima/crescimento & desenvolvimento , Hidrocefalia/patologia , Terceiro Ventrículo/fisiopatologia , Fatores Etários , Animais , Animais Recém-Nascidos , Bromodesoxiuridina/metabolismo , Contagem de Células , Proliferação de Células , Modelos Animais de Doenças , Epêndima/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Camundongos , Camundongos Mutantes Neurológicos , Microscopia Eletrônica de Varredura , Antígeno Nuclear de Célula em Proliferação/metabolismo , Terceiro Ventrículo/citologia , Tubulina (Proteína)/metabolismo
11.
J Neurosci ; 29(32): 10010-24, 2009 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-19675235

RESUMO

The region that surrounds the central canal of the spinal cord derives from the neural tube and retains a substantial degree of plasticity. In turtles, this region is a neurogenic niche where newborn neurons coexist with precursors, a fact that may be related with the endogenous repair capabilities of low vertebrates. Immunohistochemical evidence suggests that the ependyma of the mammalian spinal cord may contain cells with similar properties, but their actual nature remains unsolved. Here, we combined immunohistochemistry for cell-specific markers with patch-clamp recordings to test the hypothesis that the ependyma of neonatal rats contains immature neurons similar to those in low vertebrates. We found that a subclass of cells expressed HuC/D neuronal proteins, doublecortin, and PSA-NCAM (polysialylated neural cell adhesion molecule) but did not express NeuN (anti-neuronal nuclei). These immature neurons displayed electrophysiological properties ranging from slow Ca(2+)-mediated responses to fast repetitive Na(+) spikes, suggesting different stages of maturation. These cells originated in the embryo, because we found colocalization of neuronal markers with 5-bromo-2'-deoxyuridine when injected during embryonic day 7-17 but not in postnatal day 0-5. Our findings represent the first evidence that the ependyma of the rat spinal cord contains cells with molecular and functional features similar to immature neurons in adult neurogenic niches. The fact that these cells retain the expression of molecules that participate in migration and neuronal differentiation raises the possibility that the ependyma of the rat spinal cord is a reservoir of immature neurons in "standby mode," which under some circumstances (e.g., injury) may complete their maturation to integrate spinal circuits.


Assuntos
Epêndima/citologia , Neurônios/citologia , Medula Espinal/citologia , Ácidos/metabolismo , Potenciais de Ação , Trifosfato de Adenosina/metabolismo , Animais , Animais Recém-Nascidos , Bromodesoxiuridina , Cálcio/metabolismo , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Proteínas ELAV/metabolismo , Proteína Semelhante a ELAV 3 , Proteína Semelhante a ELAV 4 , Epêndima/crescimento & desenvolvimento , Epêndima/metabolismo , Imuno-Histoquímica , Técnicas In Vitro , Potenciais da Membrana , Proteínas Associadas aos Microtúbulos/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neurônios/metabolismo , Neurônios/ultraestrutura , Neuropeptídeos/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Ácidos Siálicos/metabolismo , Sódio/metabolismo , Medula Espinal/crescimento & desenvolvimento , Medula Espinal/metabolismo
12.
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
13.
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
14.
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
15.
Nat Neurosci ; 8(7): 865-72, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15951811

RESUMO

Adult neurogenesis in mammals is restricted to two small regions, including the olfactory bulb, where GABAergic and dopaminergic interneurons are newly generated throughout the entire lifespan. However, the mechanisms directing them towards a specific neuronal phenotype are not yet understood. Here, we demonstrate the dual role of the transcription factor Pax6 in generating neuronal progenitors and also in directing them towards a dopaminergic periglomerular phenotype in adult mice. We present further evidence that dopaminergic periglomerular neurons originate in a distinct niche, the rostral migratory stream, and are fewer derived from precursors in the zone lining the ventricle. This regionalization of the adult precursor cells is further supported by the restricted expression of the transcription factor Olig2, which specifies transit-amplifying precursor fate and opposes the neurogenic role of Pax6. Together, these data explain both extrinsic and intrinsic mechanisms controlling neuronal identity in adult neurogenesis.


Assuntos
Neurônios/fisiologia , Bulbo Olfatório/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Divisão Celular/fisiologia , Linhagem Celular/fisiologia , Dopamina/metabolismo , Epêndima/citologia , Epêndima/crescimento & desenvolvimento , Epêndima/metabolismo , Proteínas do Olho/metabolismo , Proteínas do Olho/fisiologia , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Neurônios/citologia , Bulbo Olfatório/citologia , Bulbo Olfatório/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Oligodendroglia/citologia , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados , Fenótipo , Proteínas Repressoras/metabolismo , Proteínas Repressoras/fisiologia , Células-Tronco/citologia , Distribuição Tecidual
16.
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
17.
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
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA