Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.787
Filtrar
1.
J Neuroinflammation ; 21(1): 255, 2024 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-39385253

RESUMO

Ependymal cells are arranged along the inner surfaces of the ventricles and the central canal of the spinal cord, providing anatomical, physiological and immunological barriers that maintain cerebrospinal fluid (CSF) homeostasis. Based on this, studies have found that alterations in gene expression, cell junctions, cytokine secretion and metabolic disturbances can lead to dysfunction of ependymal cells, thereby participating in the onset and progression of central nervous system (CNS) infections. Additionally, ependymal cells can exhibit proliferative and regenerative potential as well as secretory functions during CNS injury, contributing to neuroprotection and post-injury recovery. Currently, studies on ependymal cell primarily focus on the basic investigations of their morphology, function and gene expression; however, there is a notable lack of clinical translational studies examining the molecular mechanisms by which ependymal cells are involved in disease onset and progression. This limits our understanding of ependymal cells in CNS infections and the development of therapeutic applications. Therefore, this review will discuss the molecular mechanism underlying the involvement of ependymal cells in CNS infections, and explore their potential for application in clinical treatment modalities.


Assuntos
Infecções do Sistema Nervoso Central , Epêndima , Humanos , Epêndima/patologia , Epêndima/metabolismo , Animais , Infecções do Sistema Nervoso Central/terapia
2.
Acta Neuropathol Commun ; 12(1): 143, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39228008

RESUMO

Neuroepithelial tumors with fusion of PLAGL1 or amplification of PLAGL1/PLAGL2 have recently been described often with ependymoma-like or embryonal histology respectively. To further evaluate emerging entities with PLAG-family genetic alterations, the histologic, molecular, clinical, and imaging features are described for 8 clinical cases encountered at St. Jude (EWSR1-PLAGL1 fusion n = 6; PLAGL1 amplification n = 1; PLAGL2 amplification n = 1). A histologic feature observed on initial resection in a subset (4/6) of supratentorial neuroepithelial tumors with EWSR1-PLAGL1 rearrangement was the presence of concurrent ependymal and ganglionic differentiation. This ranged from prominent clusters of ganglion cells within ependymoma/subependymoma-like areas, to interspersed ganglion cells of low to moderate frequency among otherwise ependymal-like histology, or focal areas with a ganglion cell component. When present, the combination of ependymal-like and ganglionic features within a supratentorial neuroepithelial tumor may raise consideration for an EWSR1-PLAGL1 fusion, and prompt initiation of appropriate molecular testing such as RNA sequencing and methylation profiling. One of the EWSR1-PLAGL1 fusion cases showed subclonal INI1 loss in a region containing small clusters of rhabdoid/embryonal cells, and developed a prominent ganglion cell component on recurrence. As such, EWSR1-PLAGL1 neuroepithelial tumors are a tumor type in which acquired inactivation of SMARCB1 and development of AT/RT features may occur and lead to clinical progression. In contrast, the PLAGL2 and PLAGL1 amplified cases showed either embryonal histology or contained an embryonal component with a significant degree of desmin staining, which could also serve to raise consideration for a PLAG entity when present. Continued compilation of associated clinical data and histopathologic findings will be critical for understanding emerging entities with PLAG-family genetic alterations.


Assuntos
Proteína EWS de Ligação a RNA , Neoplasias Supratentoriais , Fatores de Transcrição , Humanos , Neoplasias Supratentoriais/genética , Neoplasias Supratentoriais/patologia , Feminino , Proteína EWS de Ligação a RNA/genética , Masculino , Fatores de Transcrição/genética , Criança , Neoplasias Neuroepiteliomatosas/genética , Neoplasias Neuroepiteliomatosas/patologia , Pré-Escolar , Adolescente , Adulto , Proteínas de Ligação a DNA/genética , Adulto Jovem , Diferenciação Celular , Proteínas de Fusão Oncogênica/genética , Epêndima/patologia , Rearranjo Gênico/genética , Proteínas Cromossômicas não Histona/genética
3.
Acta Neuropathol ; 148(1): 39, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39254862

RESUMO

Mature multiciliated ependymal cells line the cerebral ventricles where they form a partial barrier between the cerebrospinal fluid (CSF) and brain parenchyma and regulate local CSF microcirculation through coordinated ciliary beating. Although the ependyma is a highly specialized brain interface with barrier, trophic, and perhaps even regenerative capacity, it remains a misfit in the canon of glial neurobiology. We provide an update to seminal reviews in the field by conducting a scoping review of the post-2010 mature multiciliated ependymal cell literature. We delineate how recent findings have either called into question or substantiated classical views of the ependymal cell. Beyond this synthesis, we document the basic methodologies and study characteristics used to describe multiciliated ependymal cells since 1980. Our review serves as a comprehensive resource for future investigations of mature multiciliated ependymal cells.


Assuntos
Encéfalo , Cílios , Epêndima , Epêndima/patologia , Humanos , Animais , Cílios/patologia , Cílios/fisiologia , Encéfalo/patologia , Adulto
4.
Nature ; 634(8033): 407-414, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39294372

RESUMO

After injury, mammalian spinal cords develop scars to confine the lesion and prevent further damage. However, excessive scarring can hinder neural regeneration and functional recovery1,2. These competing actions underscore the importance of developing therapeutic strategies to dynamically modulate scar progression. Previous research on scarring has primarily focused on astrocytes, but recent evidence has suggested that ependymal cells also participate. Ependymal cells normally form the epithelial layer encasing the central canal, but they undergo massive proliferation and differentiation into astroglia following certain injuries, becoming a core scar component3-7. However, the mechanisms regulating ependymal proliferation in vivo remain unclear. Here we uncover an endogenous κ-opioid signalling pathway that controls ependymal proliferation. Specifically, we detect expression of the κ-opioid receptor, OPRK1, in a functionally under-characterized cell type known as cerebrospinal fluid-contacting neuron (CSF-cN). We also discover a neighbouring cell population that expresses the cognate ligand prodynorphin (PDYN). Whereas κ-opioids are typically considered inhibitory, they excite CSF-cNs to inhibit ependymal proliferation. Systemic administration of a κ-antagonist enhances ependymal proliferation in uninjured spinal cords in a CSF-cN-dependent manner. Moreover, a κ-agonist impairs ependymal proliferation, scar formation and motor function following injury. Together, our data suggest a paracrine signalling pathway in which PDYN+ cells tonically release κ-opioids to stimulate CSF-cNs and suppress ependymal proliferation, revealing an endogenous mechanism and potential pharmacological strategy for modulating scarring after spinal cord injury.


Assuntos
Proliferação de Células , Epêndima , Peptídeos Opioides , Transdução de Sinais , Medula Espinal , Animais , Feminino , Masculino , Camundongos , Cicatriz/metabolismo , Cicatriz/patologia , Epêndima/metabolismo , Epêndima/citologia , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Peptídeos Opioides/metabolismo , Precursores de Proteínas/metabolismo , Receptores Opioides kappa/metabolismo , Medula Espinal/metabolismo , Medula Espinal/citologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia
5.
Nat Commun ; 15(1): 6604, 2024 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-39098920

RESUMO

The ependyma lining the third ventricle (3V) in the mediobasal hypothalamus plays a crucial role in energy balance and glucose homeostasis. It is characterized by a high functional heterogeneity and plasticity, but the underlying molecular mechanisms governing its features are not fully understood. Here, 5481 hypothalamic ependymocytes were cataloged using FACS-assisted scRNAseq from fed, 12h-fasted, and 24h-fasted adult male mice. With standard clustering analysis, typical ependymal cells and ß2-tanycytes appear sharply defined, but other subpopulations, ß1- and α-tanycytes, display fuzzy boundaries with few or no specific markers. Pseudospatial approaches, based on the 3V neuroanatomical distribution, enable the identification of specific versus shared tanycyte markers and subgroup-specific versus general tanycyte functions. We show that fasting dynamically shifts gene expression patterns along the 3V, leading to a spatial redistribution of cell type-specific responses. Altogether, we show that changes in energy status induce metabolic and functional switches in tanycyte subpopulations, providing insights into molecular and functional diversity and plasticity within the tanycyte population.


Assuntos
Células Ependimogliais , Jejum , Metabolismo dos Lipídeos , Neurônios , Animais , Células Ependimogliais/metabolismo , Masculino , Jejum/metabolismo , Camundongos , Neurônios/metabolismo , Epêndima/metabolismo , Epêndima/citologia , Hipotálamo/metabolismo , Hipotálamo/citologia , Camundongos Endogâmicos C57BL , Metabolismo Energético , Terceiro Ventrículo/metabolismo , Glucose/metabolismo
6.
Cytopathology ; 35(5): 556-560, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38988178

RESUMO

Ependymal and choroid plexus tumours arise in anatomically related regions. Their intraoperative differential diagnosis is large and depends on factors such as age, tumour site and clinical presentation. Squash cytology can provide valuable information in this context. Cytological features of conventional ependymomas, subependymomas and myxopapillary ependymomas as well as choroid plexus tumours are reviewed and illustrated. Differential diagnostic considerations integrating morphological and clinical information are discussed.


Assuntos
Neoplasias do Plexo Corióideo , Ependimoma , Humanos , Neoplasias do Plexo Corióideo/patologia , Neoplasias do Plexo Corióideo/diagnóstico , Ependimoma/patologia , Ependimoma/diagnóstico , Citodiagnóstico/métodos , Diagnóstico Diferencial , Plexo Corióideo/patologia , Epêndima/patologia , Feminino
7.
Fluids Barriers CNS ; 21(1): 53, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38956598

RESUMO

AQP4 is expressed in the endfeet membranes of subpial and perivascular astrocytes and in the ependymal cells that line the ventricular system. The sporadic appearance of obstructive congenital hydrocephalus (OCHC) has been observed in the offspring of AQP4-/- mice (KO) due to stenosis of Silvio's aqueduct. Here, we explore whether the lack of AQP4 expression leads to abnormal development of ependymal cells in the aqueduct of mice. We compared periaqueductal samples from wild-type and KO mice. The microarray-based transcriptome analysis reflected a large number of genes with differential expression (809). Gene sets (GS) associated with ependymal development, ciliary function and the immune system were specially modified qPCR confirmed reduced expression in the KO mice genes: (i) coding for transcription factors for ependymal differentiation (Rfx4 and FoxJ1), (ii) involved in the constitution of the central apparatus of the axoneme (Spag16 and Hydin), (iii) associated with ciliary assembly (Cfap43, Cfap69 and Ccdc170), and (iv) involved in intercellular junction complexes of the ependyma (Cdhr4). By contrast, genes such as Spp1, Gpnmb, Itgax, and Cd68, associated with a Cd11c-positive microglial population, were overexpressed in the KO mice. Electron microscopy and Immunofluorescence of vimentin and γ-tubulin revealed a disorganized ependyma in the KO mice, with changes in the intercellular complex union, unevenly orientated cilia, and variations in the planar cell polarity of the apical membrane. These structural alterations translate into reduced cilia beat frequency, which might alter cerebrospinal fluid movement. The presence of CD11c + microglia cells in the periaqueductal zone of mice during the first postnatal week is a novel finding. In AQP4-/- mice, these cells remain present around the aqueduct for an extended period, showing peak expression at P11. We propose that these cells play an important role in the normal development of the ependyma and that their overexpression in KO mice is crucial to reduce ependyma abnormalities that could otherwise contribute to the development of obstructive hydrocephalus.


Assuntos
Aquaporina 4 , Epêndima , Hidrocefalia , Camundongos Knockout , Microglia , Animais , Epêndima/metabolismo , Epêndima/patologia , Hidrocefalia/metabolismo , Hidrocefalia/genética , Hidrocefalia/patologia , Microglia/metabolismo , Aquaporina 4/metabolismo , Aquaporina 4/genética , Camundongos , Aqueduto do Mesencéfalo/metabolismo , Aqueduto do Mesencéfalo/patologia , Antígenos CD11/metabolismo , Antígenos CD11/genética , Camundongos Endogâmicos C57BL
8.
Biol Pharm Bull ; 47(6): 1113-1118, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38839362

RESUMO

Motile cilia in the ependymal cells that line the brain ventricles play pivotal roles in cerebrospinal fluid (CSF) flow in well-defined directions. However, the substances and pathways which regulate their beating have not been well studied. Here, we used primary cultured cells derived from neonatal mouse brain that possess motile cilia and found that adenosine (ADO) stimulates ciliary beating by increasing the ciliary beat frequency (CBF) in a concentration-dependent manner, with the ED50 value being 5 µM. Ciliary beating stimulated by ADO was inhibited by A2B receptor (A2BR) antagonist MRS1754 without any inhibition by antagonists of other ADO receptor subtypes. The expression of A2BR on the cilia was also confirmed by immunofluorescence. The values of CBF were also increased by forskolin, which is an activator of adenylate cyclase, whereas they were not further increased by the addition of ADO. Furthermore, ciliary beating was not stimulated by ADO in the presence of a protein kinase A (PKA) inhibitors. These results altogether suggest that ADO stimulates ciliary beating through A2BR on the cilia, and activation of PKA.


Assuntos
Adenosina , Animais Recém-Nascidos , Encéfalo , Cílios , Proteínas Quinases Dependentes de AMP Cíclico , Receptor A2B de Adenosina , Animais , Cílios/efeitos dos fármacos , Cílios/metabolismo , Cílios/fisiologia , Receptor A2B de Adenosina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Adenosina/farmacologia , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Camundongos , Células Cultivadas , Transdução de Sinais/efeitos dos fármacos , Antagonistas do Receptor A2 de Adenosina/farmacologia , Colforsina/farmacologia , Epêndima/metabolismo , Epêndima/citologia
9.
Brain Res ; 1840: 149082, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-38866307

RESUMO

Ubiquitin specific protease 18 (USP18) serves as a potent inhibitor of Type I interferon (IFN) signaling. Previous studies have shown that Usp18 deficient (homozygous Usp18 gene knockout) mice exhibit hydrocephalus; however, the precise molecular mechanism underlying hydrocephalus development remains elusive. In this study, we demonstrate that mice lacking both type I IFN receptor subunit 1 (Ifnar1) and Usp18 (Ifnar1/Usp18 double knockout mice) are viable and do not display a hydrocephalus phenotype. Moreover, we observed that suppression of USP18 in ependymal cells treated with IFN significantly increased cell death, including pyroptosis, and decreased proliferation. These findings suggest that heightened sensitivity to type I IFN during brain development contributes to the onset of hydrocephalus. Furthermore, they imply that inhibition of IFN signaling may hold promise as a therapeutic strategy for hydrocephalus.


Assuntos
Hidrocefalia , Interferon Tipo I , Camundongos Knockout , Receptor de Interferon alfa e beta , Ubiquitina Tiolesterase , Animais , Hidrocefalia/genética , Hidrocefalia/patologia , Interferon Tipo I/metabolismo , Camundongos , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/efeitos dos fármacos , Epêndima/metabolismo , Proliferação de Células/efeitos dos fármacos , Piroptose/efeitos dos fármacos , Piroptose/fisiologia
10.
J Neurosci Methods ; 409: 110198, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38878975

RESUMO

BACKGROUND: Ependymal cilia play a major role in the circulation of cerebrospinal fluid. Although isolation of cilia is an essential technique for investigating ciliary structure, to the best of our knowledge, no report on the isolation and structural analysis of ependymal cilia from mouse brain is available. NEW METHOD: We developed a novel method for isolating ependymal cilia from mouse brain ventricles. We isolated ependymal cilia by partially opening the lateral ventricles and gently applying shear stress, followed by pipetting and ultracentrifugation. RESULTS: Using this new method, we were able to observe cilia separately. The results demonstrated that our method successfully isolated intact ependymal cilia with preserved morphology and ultrastructure. In this procedure, the ventricular ependymal cell layer was partially detached. COMPARISON WITH EXISTING METHODS: Compared to existing methods for isolating cilia from other tissues, our method is meticulously tailored for extracting ependymal cilia from the mouse brain. Designed with a keen understanding of the fragility of the ventricular ependyma, our method prioritizes minimizing tissue damage during the isolation procedure. CONCLUSIONS: We isolated ependymal cilia from mouse brain by applying shear stress selectively to the ventricles. Our method can be used to conduct more detailed studies on the structure of ependymal cilia.


Assuntos
Cílios , Epêndima , Animais , Cílios/fisiologia , Epêndima/citologia , Camundongos , Camundongos Endogâmicos C57BL , Encéfalo/citologia , Masculino , Ventrículos Cerebrais/citologia
11.
Proc Natl Acad Sci U S A ; 121(27): e2314702121, 2024 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-38916997

RESUMO

Enlargement of the cerebrospinal fluid (CSF)-filled brain ventricles (cerebral ventriculomegaly), the cardinal feature of congenital hydrocephalus (CH), is increasingly recognized among patients with autism spectrum disorders (ASD). KATNAL2, a member of Katanin family microtubule-severing ATPases, is a known ASD risk gene, but its roles in human brain development remain unclear. Here, we show that nonsense truncation of Katnal2 (Katnal2Δ17) in mice results in classic ciliopathy phenotypes, including impaired spermatogenesis and cerebral ventriculomegaly. In both humans and mice, KATNAL2 is highly expressed in ciliated radial glia of the fetal ventricular-subventricular zone as well as in their postnatal ependymal and neuronal progeny. The ventriculomegaly observed in Katnal2Δ17 mice is associated with disrupted primary cilia and ependymal planar cell polarity that results in impaired cilia-generated CSF flow. Further, prefrontal pyramidal neurons in ventriculomegalic Katnal2Δ17 mice exhibit decreased excitatory drive and reduced high-frequency firing. Consistent with these findings in mice, we identified rare, damaging heterozygous germline variants in KATNAL2 in five unrelated patients with neurosurgically treated CH and comorbid ASD or other neurodevelopmental disorders. Mice engineered with the orthologous ASD-associated KATNAL2 F244L missense variant recapitulated the ventriculomegaly found in human patients. Together, these data suggest KATNAL2 pathogenic variants alter intraventricular CSF homeostasis and parenchymal neuronal connectivity by disrupting microtubule dynamics in fetal radial glia and their postnatal ependymal and neuronal descendants. The results identify a molecular mechanism underlying the development of ventriculomegaly in a genetic subset of patients with ASD and may explain persistence of neurodevelopmental phenotypes in some patients with CH despite neurosurgical CSF shunting.


Assuntos
Cílios , Hidrocefalia , Microtúbulos , Animais , Feminino , Humanos , Masculino , Camundongos , ATPases Associadas a Diversas Atividades Celulares/genética , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/patologia , Transtorno do Espectro Autista/metabolismo , Cílios/metabolismo , Cílios/patologia , Epêndima/metabolismo , Epêndima/patologia , Hidrocefalia/genética , Hidrocefalia/patologia , Hidrocefalia/metabolismo , Katanina/metabolismo , Katanina/genética , Microtúbulos/metabolismo , Neurônios/metabolismo , Células Piramidais/metabolismo , Células Piramidais/patologia
12.
J Neurochem ; 168(10): 3449-3466, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38702968

RESUMO

Ependymal cells form a specialized brain-cerebrospinal fluid (CSF) interface and regulate local CSF microcirculation. It is becoming increasingly recognized that ependymal cells assume a reactive state in response to aging and disease, including conditions involving hypoxia, hydrocephalus, neurodegeneration, and neuroinflammation. Yet what transcriptional signatures govern these reactive states and whether this reactivity shares any similarities with classical descriptions of glial reactivity (i.e., in astrocytes) remain largely unexplored. Using single-cell transcriptomics, we interrogated this phenomenon by directly comparing the reactive ependymal cell transcriptome to the reactive astrocyte transcriptome using a well-established model of autoimmune-mediated neuroinflammation (MOG35-55 EAE). In doing so, we unveiled core glial reactivity-associated genes that defined the reactive ependymal cell and astrocyte response to MOG35-55 EAE. Interestingly, known reactive astrocyte genes from other CNS injury/disease contexts were also up-regulated by MOG35-55 EAE ependymal cells, suggesting that this state may be conserved in response to a variety of pathologies. We were also able to recapitulate features of the reactive ependymal cell state acutely using a classic neuroinflammatory cocktail (IFNγ/LPS) both in vitro and in vivo. Taken together, by comparing reactive ependymal cells and astrocytes, we identified a conserved signature underlying glial reactivity that was present in several neuroinflammatory contexts. Future work will explore the mechanisms driving ependymal reactivity and assess downstream functional consequences.


Assuntos
Astrócitos , Encefalomielite Autoimune Experimental , Epêndima , Camundongos Endogâmicos C57BL , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Epêndima/metabolismo , Epêndima/patologia , Camundongos , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Feminino , Doenças Neuroinflamatórias/patologia , Transcriptoma
13.
PLoS Biol ; 22(5): e3002596, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38718086

RESUMO

Autism spectrum disorders (ASD) frequently accompany macrocephaly, which often involves hydrocephalic enlargement of brain ventricles. Katnal2 is a microtubule-regulatory protein strongly linked to ASD, but it remains unclear whether Katnal2 knockout (KO) in mice leads to microtubule- and ASD-related molecular, synaptic, brain, and behavioral phenotypes. We found that Katnal2-KO mice display ASD-like social communication deficits and age-dependent progressive ventricular enlargements. The latter involves increased length and beating frequency of motile cilia on ependymal cells lining ventricles. Katnal2-KO hippocampal neurons surrounded by enlarged lateral ventricles show progressive synaptic deficits that correlate with ASD-like transcriptomic changes involving synaptic gene down-regulation. Importantly, early postnatal Katnal2 re-expression prevents ciliary, ventricular, and behavioral phenotypes in Katnal2-KO adults, suggesting a causal relationship and a potential treatment. Therefore, Katnal2 negatively regulates ependymal ciliary function and its deletion in mice leads to ependymal ciliary hyperfunction and hydrocephalus accompanying ASD-related behavioral, synaptic, and transcriptomic changes.


Assuntos
Transtorno do Espectro Autista , Cílios , Epêndima , Camundongos Knockout , Fenótipo , Animais , Masculino , Camundongos , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/fisiopatologia , Comportamento Animal , Cílios/metabolismo , Modelos Animais de Doenças , Epêndima/metabolismo , Hipocampo/metabolismo , Hidrocefalia/genética , Hidrocefalia/metabolismo , Hidrocefalia/patologia , Hidrocefalia/fisiopatologia , Katanina/metabolismo , Katanina/genética , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Sinapses/metabolismo , Transcriptoma/genética
14.
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
15.
Tissue Eng Regen Med ; 21(4): 625-639, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38578425

RESUMO

BACKGROUND: Syringomyelia is a progressive chronic disease that leads to nerve pain, sensory dissociation, and dyskinesia. Symptoms often do not improve after surgery. Stem cells have been widely explored for the treatment of nervous system diseases due to their immunoregulatory and neural replacement abilities. METHODS: In this study, we used a rat model of syringomyelia characterized by focal dilatation of the central canal to explore an effective transplantation scheme and evaluate the effect of mesenchymal stem cells and induced neural stem cells for the treatment of syringomyelia. RESULTS: The results showed that cell transplantation could not only promote syrinx shrinkage but also stimulate the proliferation of ependymal cells, and the effect of this result was related to the transplantation location. These reactions appeared only when the cells were transplanted into the cavity. Additionally, we discovered that cell transplantation transformed activated microglia into the M2 phenotype. IGF1-expressing M2 microglia may play a significant role in the repair of nerve pain. CONCLUSION: Cell transplantation can promote cavity shrinkage and regulate the local inflammatory environment. Moreover, the proliferation of ependymal cells may indicate the activation of endogenous stem cells, which is important for the regeneration and repair of spinal cord injury.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Células-Tronco Neurais , Ratos Sprague-Dawley , Siringomielia , Animais , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/citologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Siringomielia/terapia , Ratos , Proliferação de Células , Epêndima , Masculino , Microglia/metabolismo , Modelos Animais de Doenças
16.
Stroke ; 55(4): 1062-1074, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38436063

RESUMO

BACKGROUND: In preterm birth germinal matrix hemorrhages (GMHs) and the consequent posthemorrhagic hydrocephalus (PHH), the neuroepithelium/ependyma development is disrupted. This work is aimed to explore the possibilities of ependymal repair in GMH/PHH using a combination of neural stem cells, ependymal progenitors (EpPs), and mesenchymal stem cells. METHODS: GMH/PHH was induced in 4-day-old mice using collagenase, blood, or blood serum injections. PHH severity was characterized 2 weeks later using magnetic resonance, immunofluorescence, and protein expression quantification with mass spectrometry. Ependymal restoration and wall regeneration after stem cell treatments were tested in vivo and in an ex vivo experimental approach using ventricular walls from mice developing moderate and severe GMH/PHH. The effect of the GMH environment on EpP differentiation was tested in vitro. Two-tailed Student t or Wilcoxon-Mann-Whitney U test was used to find differences between the treated and nontreated groups. ANOVA and Kruskal-Wallis tests were used to compare >2 groups with post hoc Tukey and Dunn multiple comparison tests, respectively. RESULTS: PHH severity was correlated with the extension of GMH and ependymal disruption (means, 88.22% severe versus 19.4% moderate). GMH/PHH hindered the survival rates of the transplanted neural stem cells/EpPs. New multiciliated ependymal cells could be generated from transplanted neural stem cells and more efficiently from EpPs (15% mean increase). Blood and TNFα (tumor necrosis factor alpha) negatively affected ciliogenesis in cells committed to ependyma differentiation (expressing Foxj1 [forkhead box J1] transcription factor). Pretreatment with mesenchymal stem cells improved the survival rates of EpPs and ependymal differentiation while reducing the edematous (means, 18% to 0.5% decrease in severe edema) and inflammatory conditions in the explants. The effectiveness of this therapeutical strategy was corroborated in vivo (means, 29% to 0% in severe edema). CONCLUSIONS: In GMH/PHH, the ependyma can be restored and edema decreased from either neural stem cell or EpP transplantation in vitro and in vivo. Mesenchymal stem cell pretreatment improved the success of the ependymal restoration.


Assuntos
Doenças Fetais , Hidrocefalia , Células-Tronco Neurais , Nascimento Prematuro , Humanos , Feminino , Animais , Camundongos , Epêndima/patologia , Hidrocefalia/cirurgia , Hidrocefalia/metabolismo , Hemorragia Cerebral/terapia , Hemorragia Cerebral/metabolismo , Edema
17.
Neurochirurgie ; 70(4): 101553, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38547561

RESUMO

BACKGROUND: Ependymal cysts (EC) typically present supra-tentorially near the lateral ventricle, juxta ventricular, or temporoparietal regions. Previous cases have also identified infratentorial EC of the brainstem, cerebellum, and subarachnoid spaces. They are mostly asymptomatic. In this paper, we present the first-ever case of a symptomatic medullary ependymal cyst treated with surgery, along with a comprehensive review of the literature on EC of other parts of the brain stem. CASE DESCRIPTION: This 51-year-old female presented with hearing loss, dizziness, diplopia, and ataxia. Radiographic imaging indicated the presence of a non-enhancing lesion in the medulla with a mass effect on the brainstem. Pathological examination confirmed its characterization as an ependymal cyst. The patient underwent a suboccipital craniotomy for the fenestration of the medullary ependymal cyst under neuro-navigation, Intra-op ultrasound and intra-operative neuro-monitoring. Histopathological examination confirmed the diagnosis of an ependymal cyst. At one month follow-up, her KPS is 90, ECOG PS 1, and her ataxia has improved with complete resolution of diplopia. CONCLUSION: Due to their rarity and potential similarity to other cystic structures, EC may be overlooked or incorrectly diagnosed resulting in mismanagement and surgical disaster. Therefore, a comprehensive understanding and awareness of their distinct characteristics are essential for accurate diagnosis and appropriate management.


Assuntos
Bulbo , Humanos , Feminino , Pessoa de Meia-Idade , Bulbo/cirurgia , Bulbo/patologia , Epêndima/cirurgia , Epêndima/patologia , Cistos do Sistema Nervoso Central/cirurgia , Cistos do Sistema Nervoso Central/diagnóstico , Neoplasias do Tronco Encefálico/cirurgia , Neoplasias do Tronco Encefálico/diagnóstico , Neoplasias Infratentoriais/cirurgia , Neoplasias Infratentoriais/diagnóstico , Imageamento por Ressonância Magnética , Craniotomia/métodos
18.
Dis Model Mech ; 17(2)2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38235522

RESUMO

Motile cilia on ependymal cells that line brain ventricular walls beat in concert to generate a flow of laminar cerebrospinal fluid (CSF). Dyneins and kinesins are ATPase microtubule motor proteins that promote the rhythmic beating of cilia axonemes. Despite common consensus about the importance of axonemal dynein motor proteins, little is known about how kinesin motors contribute to cilia motility. Here, we show that Kif6 is a slow processive motor (12.2±2.0 nm/s) on microtubules in vitro and localizes to both the apical cytoplasm and the axoneme in ependymal cells, although it does not display processive movement in vivo. Using a mouse mutant that models a human Kif6 mutation in a proband displaying macrocephaly, hypotonia and seizures, we found that loss of Kif6 function causes decreased ependymal cilia motility and, subsequently, decreases fluid flow on the surface of brain ventricular walls. Disruption of Kif6 also disrupts orientation of cilia, formation of robust apical actin networks and stabilization of basal bodies at the apical surface. This suggests a role for the Kif6 motor protein in the maintenance of ciliary homeostasis within ependymal cells.


Assuntos
Cílios , Cinesinas , Humanos , Encéfalo/metabolismo , Cílios/metabolismo , Dineínas/metabolismo , Epêndima , Cinesinas/metabolismo
19.
J Anat ; 244(3): 391-401, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-37965891

RESUMO

In humans and most mammals, there is a notch-like portal, the foramen of Luschka (or lateral foramen), which connects the lumen of the fourth ventricle with the subdural space. Gross dissection, light and scanning electron microscopy, and µCT analysis revealed the presence of a foramen of Luschka in the American alligator (Alligator mississippiensis). In this species, the foramen of Luschka is a notch in the dorsolateral wall of the pons immediately caudal to the peduncular base of the cerebellum, near the rostral end of the telovelar membrane over the fourth ventricle. At the foramen of Luschka there was a transition from a superficial pia mater lining to a deep ependymal lining. There was continuity between the lumen of the fourth ventricle and the subdural space, via the foramen of Luschka. This anatomical continuity was further demonstrated by injecting Evans blue into the lateral ventricle which led to extravasation through the foramen of Luschka and pooling of the dye on the lateral surface of the brain. Simultaneous subdural and intraventricular recordings of cerebrospinal fluid (CSF) pressures revealed a stable agreement between the two pressures at rest. Perturbation of the system allowed for static and dynamic differences to develop, which could indicate varying flow patterns of CSF through the foramen of Luschka.


Assuntos
Jacarés e Crocodilos , Animais , Humanos , Espaço Subdural , Cerebelo , Quarto Ventrículo , Epêndima , Mamíferos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA