Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 511
Filter
1.
Stroke ; 55(4): 1062-1074, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38436063

ABSTRACT

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.


Subject(s)
Fetal Diseases , Hydrocephalus , Neural Stem Cells , Premature Birth , Humans , Female , Animals , Mice , Ependyma/pathology , Hydrocephalus/surgery , Hydrocephalus/metabolism , Cerebral Hemorrhage/therapy , Cerebral Hemorrhage/metabolism , Edema
3.
Methods Cell Biol ; 176: 103-123, 2023.
Article in English | MEDLINE | ID: mdl-37164533

ABSTRACT

Cilia are well conserved hair-like structures that have diverse sensory and motile functions. In the brain, motile ciliated cells, known as ependymal cells, line the cerebrospinal fluid (CSF) filled ventricles, where their beating contribute to fluid movement. Ependymal cells have gathered increasing interest since they are associated with hydrocephalus, a neurological condition with ventricular enlargement. In this article, we highlight methods to identify and characterize motile ciliated ependymal lineage in the brain of zebrafish using histological staining and transgenic reporter lines.


Subject(s)
Hydrocephalus , Zebrafish , Animals , Zebrafish/genetics , Brain/pathology , Ependyma/metabolism , Ependyma/pathology , Hydrocephalus/genetics , Hydrocephalus/metabolism , Hydrocephalus/pathology , Animals, Genetically Modified , Cilia/metabolism
4.
Biomolecules ; 13(5)2023 04 27.
Article in English | MEDLINE | ID: mdl-37238624

ABSTRACT

The neuron loss caused by the progressive damage to the nervous system is proposed to be the main pathogenesis of neurodegenerative diseases. Ependyma is a layer of ciliated ependymal cells that participates in the formation of the brain-cerebrospinal fluid barrier (BCB). It functions to promotes the circulation of cerebrospinal fluid (CSF) and the material exchange between CSF and brain interstitial fluid. Radiation-induced brain injury (RIBI) shows obvious impairments of the blood-brain barrier (BBB). In the neuroinflammatory processes after acute brain injury, a large amount of complement proteins and infiltrated immune cells are circulated in the CSF to resist brain damage and promote substance exchange through the BCB. However, as the protective barrier lining the brain ventricles, the ependyma is extremely vulnerable to cytotoxic and cytolytic immune responses. When the ependyma is damaged, the integrity of BCB is destroyed, and the CSF flow and material exchange is affected, leading to brain microenvironment imbalance, which plays a vital role in the pathogenesis of neurodegenerative diseases. Epidermal growth factor (EGF) and other neurotrophic factors promote the differentiation and maturation of ependymal cells to maintain the integrity of the ependyma and the activity of ependymal cilia, and may have therapeutic potential in restoring the homeostasis of the brain microenvironment after RIBI or during the pathogenesis of neurodegenerative diseases.


Subject(s)
Brain Injuries , Neurodegenerative Diseases , Humans , Ependyma/metabolism , Ependyma/pathology , Nerve Growth Factors/metabolism , Neurodegenerative Diseases/metabolism , Brain/metabolism , Brain Injuries/metabolism
5.
PLoS Biol ; 21(3): e3002008, 2023 03.
Article in English | MEDLINE | ID: mdl-36862758

ABSTRACT

Idiopathic scoliosis (IS) is the most common spinal deformity diagnosed in childhood or early adolescence, while the underlying pathogenesis of this serious condition remains largely unknown. Here, we report zebrafish ccdc57 mutants exhibiting scoliosis during late development, similar to that observed in human adolescent idiopathic scoliosis (AIS). Zebrafish ccdc57 mutants developed hydrocephalus due to cerebrospinal fluid (CSF) flow defects caused by uncoordinated cilia beating in ependymal cells. Mechanistically, Ccdc57 localizes to ciliary basal bodies and controls the planar polarity of ependymal cells through regulating the organization of microtubule networks and proper positioning of basal bodies. Interestingly, ependymal cell polarity defects were first observed in ccdc57 mutants at approximately 17 days postfertilization, the same time when scoliosis became apparent and prior to multiciliated ependymal cell maturation. We further showed that mutant spinal cord exhibited altered expression pattern of the Urotensin neuropeptides, in consistent with the curvature of the spine. Strikingly, human IS patients also displayed abnormal Urotensin signaling in paraspinal muscles. Altogether, our data suggest that ependymal polarity defects are one of the earliest sign of scoliosis in zebrafish and disclose the essential and conserved roles of Urotensin signaling during scoliosis progression.


Subject(s)
Hydrocephalus , Scoliosis , Urotensins , Animals , Cilia/metabolism , Ependyma/metabolism , Ependyma/pathology , Hydrocephalus/genetics , Hydrocephalus/metabolism , Hydrocephalus/pathology , Scoliosis/genetics , Scoliosis/metabolism , Scoliosis/pathology , Urotensins/metabolism , Zebrafish
6.
Neurobiol Dis ; 175: 105913, 2022 12.
Article in English | MEDLINE | ID: mdl-36341771

ABSTRACT

Dysfunction of motile cilia in ependymal cells has been proposed to be a pathogenic cause of cerebrospinal fluid (CSF) overaccumulation leading to ventricular expansion in hydrocephalus, primarily based on observations of enlarged ventricles in mouse models of primary ciliary dyskinesia. Here, we review human and animal evidence that warrants a rethinking of the cilia hypothesis in hydrocephalus. First, we discuss neuroembryology and physiology data that do not support a role for ependymal cilia as the primary propeller of CSF movement across the ventricles in the human brain, particularly during in utero development prior to the functional maturation of ependymal cilia. Second, we highlight that in contrast to mouse models, motile ciliopathies infrequently cause hydrocephalus in humans. Instead, gene mutations affecting motile cilia function impact not only ependymal cilia but also motile cilia found in other organ systems outside of the brain, causing a clinical syndrome of recurrent respiratory infections and situs inversus, symptoms that do not typically accompany most cases of human hydrocephalus. Finally, we postulate that certain cases of hydrocephalus associated with ciliary gene mutations may arise not necessarily just from loss of cilia-generated CSF flow but also from altered neurodevelopment, given the potential functions of ciliary genes in signaling and neural stem cell fate beyond generating fluid flow. Further investigations are needed to clarify the link between motile cilia, CSF physiology, and brain development, the understanding of which has implications for the care of patients with hydrocephalus and other related neurodevelopmental disorders.


Subject(s)
Cilia , Hydrocephalus , Animals , Mice , Humans , Cilia/pathology , Hydrocephalus/etiology , Hydrocephalus/pathology , Ependyma/pathology , Brain/pathology , Disease Models, Animal
7.
Br J Neurosurg ; 36(3): 329-339, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35579079

ABSTRACT

Congenital hydrocephalus affects approximately one in 1000 newborn children and is fatal in approximately 50% of untreated cases. The currently known management protocols usually necessitate multiple interventions and long-term use of healthcare resources due to a relatively high incidence of complications, and many of them mostly provide a treatment of the effect rather than the cause of cerebrospinal fluid flow reduction or outflow obstruction. Future studies discussing etiology specific hydrocephalus alternative treatments are needed. We systematically reviewed the available literature on the effect of ciliary abnormality on congenital hydrocephalus pathogenesis, to open a discussion on the feasibility of factoring ciliary abnormality in future research on hydrocephalus treatment modalities. Although there are different forms of ciliopathies, we focused in this review on primary ciliary dyskinesia. There is growing evidence of association of other ciliary syndromes and hydrocephalus, such as the reduced generation of multiple motile cilia, which is distinct from primary ciliary dyskinesia. Data for this review were identified by searching PubMed using the search terms 'hydrocephalus,' 'Kartagener syndrome,' 'primary ciliary dyskinesia,' and 'immotile cilia syndrome.' Only articles published in English and reporting human patients were included. Seven studies met our inclusion criteria, reporting 12 cases of hydrocephalus associated with primary ciliary dyskinesia. The patients had variable clinical presentations, genetic backgrounds, and ciliary defects. The ependymal water propelling cilia differ in structure and function from the mucus propelling cilia, and there is a possibility of isolated non-syndromic ependymal ciliopathy causing only hydrocephalus with growing evidence in the literature for the association ependymal ciliary abnormality and hydrocephalus. Abdominal and thoracic situs in children with hydrocephalus can be evaluated, and secondary damage of ependymal cilia causing hydrocephalus in cases with generalized ciliary abnormality can be considered.


Subject(s)
Hydrocephalus , Kartagener Syndrome , Cilia/genetics , Cilia/pathology , Ependyma/pathology , Humans , Hydrocephalus/etiology , Hydrocephalus/pathology , Infant, Newborn , Kartagener Syndrome/complications , Kartagener Syndrome/genetics , Kartagener Syndrome/pathology
8.
Neurosci Res ; 181: 22-38, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35452717

ABSTRACT

The adult mammalian central nervous system has limited regenerative ability, and spinal cord injury (SCI) often causes lifelong motor disability. While regeneration is limited in adults, injured spinal cord tissue can be regenerated and neural function can be almost completely restored in neonates. However, difference of cellular composition in lesion has not been well characterized. To gain insight into the age-dependent cellular reaction after SCI, we performed single-nucleus RNA sequencing, analyzing 4076 nuclei from sham and injured spinal cords from adult and neonatal mice. Clustering analysis identified 18 cell populations. We identified previously undescribed cells with ependymal cell-like gene expression profile, the number of which was increased in neonates after SCI. Histological analysis revealed that these cells line the central canal under physiological conditions in both adults and neonates. We confirmed that they were enriched in the lesion only in neonates. We further showed that these cells were positive for the cellular markers of ependymal cells, astrocytes and radial glial cells. This study provides a deeper understanding of neonate-specific cellular responses after SCI, which may determine regenerative capacity.


Subject(s)
Disabled Persons , Motor Disorders , Spinal Cord Injuries , Animals , Animals, Newborn , Ependyma/metabolism , Ependyma/pathology , Humans , Mammals , Mice , Motor Disorders/metabolism , Sequence Analysis, RNA , Spinal Cord/metabolism , Spinal Cord Injuries/pathology
9.
J Clin Neurosci ; 99: 10-16, 2022 May.
Article in English | MEDLINE | ID: mdl-35228087

ABSTRACT

BACKGROUND: Intracranial ependymal cysts (IECs) are rare, histologically benign neuroepithelial cysts that mostly occur in the cerebral parenchyma. The majority of these cysts are clinically silent and discovered incidentally, but when symptomatic they may compress surrounding structures, thus surgical intervention is needed. The current data in the literature about ECs is very scarce, and many are misdiagnosed, once they share many radiological characteristics with a variety of intracranial benign cysts. Also their terminology is confusing, and its definitive diagnosis can only be made through a thorough histopathological study, hence a detailed description about these uncommon lesions is necessary. The correct identification of the lesion lead to our better understanding of the condition and further improvement of the patient's prognosis. METHODS: A descriptive case is presented; moreover, a detailed PubMed search according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline was performed. The data found was analyzed by various criteria in order to correctly describe the characteristics of this lesion. RESULTS: The literature review gathered 9 descriptions of patients with IECs with a diverse range anatomopathological and clinical manifestations. All of the included studies found were case reports. Moreover, the authors suggest an updated classification of the lesion, involving their immunohistochemical characteristics. CONCLUSIONS: The information obtained from this study highlights IECs rarity and their inaccurately classification. We propose that the definitive diagnosis of IECs shall be made upon histopathological confirmation of an ependyma-lined cyst along with a positive glial fibrillary acidic protein (GFAP).


Subject(s)
Central Nervous System Cysts , Cysts , Central Nervous System Cysts/diagnostic imaging , Central Nervous System Cysts/pathology , Cysts/diagnostic imaging , Cysts/pathology , Ependyma/pathology , Ependyma/surgery , Humans
10.
Sci Rep ; 11(1): 21956, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34753951

ABSTRACT

Deep and periventricular white matter hyperintensities (dWMH/pvWMH) are bright appearing white matter tissue lesions in T2-weighted fluid attenuated inversion recovery magnetic resonance images and are frequent observations in the aging human brain. While early stages of these white matter lesions are only weakly associated with cognitive impairment, their progressive growth is a strong indicator for long-term functional decline. DWMHs are typically associated with vascular degeneration in diffuse white matter locations; for pvWMHs, however, no unifying theory exists to explain their consistent onset around the horns of the lateral ventricles. We use patient imaging data to create anatomically accurate finite element models of the lateral ventricles, white and gray matter, and cerebrospinal fluid, as well as to reconstruct their WMH volumes. We simulated the mechanical loading of the ependymal cells forming the primary brain-fluid interface, the ventricular wall, and its surrounding tissues at peak ventricular pressure during the hemodynamic cycle. We observe that both the maximum principal tissue strain and the largest ependymal cell stretch consistently localize in the anterior and posterior horns. Our simulations show that ependymal cells experience a loading state that causes the ventricular wall to be stretched thin. Moreover, we show that maximum wall loading coincides with the pvWMH locations observed in our patient scans. These results warrant further analysis of white matter pathology in the periventricular zone that includes a mechanics-driven deterioration model for the ventricular wall.


Subject(s)
Ependyma/pathology , White Matter/pathology , Aged , Ependyma/diagnostic imaging , Female , Humans , Lateral Ventricles/diagnostic imaging , Magnetic Resonance Imaging/methods , Male , White Matter/diagnostic imaging
11.
Nat Commun ; 12(1): 5887, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34620853

ABSTRACT

TRIP6, a member of the ZYXIN-family of LIM domain proteins, is a focal adhesion component. Trip6 deletion in the mouse, reported here, reveals a function in the brain: ependymal and choroid plexus epithelial cells are carrying, unexpectedly, fewer and shorter cilia, are poorly differentiated, and the mice develop hydrocephalus. TRIP6 carries numerous protein interaction domains and its functions require homodimerization. Indeed, TRIP6 disruption in vitro (in a choroid plexus epithelial cell line), via RNAi or inhibition of its homodimerization, confirms its function in ciliogenesis. Using super-resolution microscopy, we demonstrate TRIP6 localization at the pericentriolar material and along the ciliary axoneme. The requirement for homodimerization which doubles its interaction sites, its punctate localization along the axoneme, and its co-localization with other cilia components suggest a scaffold/co-transporter function for TRIP6 in cilia. Thus, this work uncovers an essential role of a LIM-domain protein assembly factor in mammalian ciliogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Brain/metabolism , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Brain/pathology , Ependyma/pathology , Focal Adhesions/metabolism , Gene Expression Regulation , Mice , Mice, Knockout , RNA Interference , Transcriptome
12.
Cell Rep ; 37(1): 109775, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34610312

ABSTRACT

Motile cilia defects impair cerebrospinal fluid (CSF) flow and can cause brain and spine disorders. The development of ciliated cells, their impact on CSF flow, and their function in brain and axial morphogenesis are not fully understood. We have characterized motile ciliated cells within the zebrafish brain ventricles. We show that the ventricles undergo restructuring through development, involving a transition from mono- to multiciliated cells (MCCs) driven by gmnc. MCCs co-exist with monociliated cells and generate directional flow patterns. These ciliated cells have different developmental origins and are genetically heterogenous with respect to expression of the Foxj1 family of ciliary master regulators. Finally, we show that cilia loss from the tela choroida and choroid plexus or global perturbation of multiciliation does not affect overall brain or spine morphogenesis but results in enlarged ventricles. Our findings establish that motile ciliated cells are generated by complementary and sequential transcriptional programs to support ventricular development.


Subject(s)
Brain/metabolism , Cilia/metabolism , Ependyma/metabolism , Animals , Animals, Genetically Modified/metabolism , Brain/cytology , Brain/pathology , Cell Lineage , Cerebrospinal Fluid/physiology , Cilia/pathology , Embryo, Nonmammalian/metabolism , Ependyma/cytology , Ependyma/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Editing , Morphogenesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Spine/growth & development , Spine/metabolism , Telencephalon/cytology , Telencephalon/metabolism , Telencephalon/pathology , Tubulin/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
Cell Death Dis ; 12(8): 766, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344859

ABSTRACT

Ependymal cells have been suggested to act as neural stem cells and exert beneficial effects after spinal cord injury (SCI). However, the molecular mechanism underlying ependymal cell regulation after SCI remains unknown. To examine the possible effect of IL-17A on ependymal cell proliferation after SCI, we locally administrated IL-17A neutralizing antibody to the injured spinal cord of a contusion SCI mouse model, and revealed that IL-17A neutralization promoted ependymal cell proliferation, which was paralleled by functional recovery and axonal reorganization of both the corticospinal tract and the raphespinal tract. Further, to test whether ependymal cell-specific manipulation of IL-17A signaling is enough to affect the outcomes of SCI, we generated ependymal cell-specific conditional IL-17RA-knockout mice and analyzed their anatomical and functional response to SCI. As a result, conditional knockout of IL-17RA in ependymal cells enhanced both axonal growth and functional recovery, accompanied by an increase in mRNA expression of neurotrophic factors. Thus, Ependymal cells may enhance the regenerative process partially by secreting neurotrophic factors, and IL-17A stimulation negatively regulates this beneficial effect. Molecular manipulation of ependymal cells might be a viable strategy for improving functional recovery.


Subject(s)
Ependyma/pathology , Interleukin-17/metabolism , Recovery of Function , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Animals , Antibodies, Neutralizing/pharmacology , Behavior, Animal/drug effects , Cell Proliferation/drug effects , Female , Interleukin-17/genetics , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Nerve Growth Factors/metabolism , Neurogenesis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-17/metabolism , Recovery of Function/drug effects , Signal Transduction , Tamoxifen/administration & dosage , Tamoxifen/pharmacology , Up-Regulation/drug effects
14.
J Neurosci Res ; 99(10): 2592-2609, 2021 10.
Article in English | MEDLINE | ID: mdl-34288039

ABSTRACT

The cytoskeleton of ependymal cells is fundamental to organize and maintain the normal architecture of the central canal (CC). However, little is known about the plasticity of cytoskeletal components after spinal cord injury. Here, we focus on the structural organization of the cytoskeleton of ependymal cells in the normal and injured spinal cord of mice (both females and males) using immunohistochemical and electron microscopy techniques. We found that in uninjured animals, the actin cytoskeleton (as revealed by phalloidin staining) was arranged following the typical pattern of polarized epithelial cells with conspicuous actin pools located in the apical domain of ependymal cells. Transmission electron microscopy images showed microvilli tufts, long cilia, and characteristic intercellular membrane specializations. After spinal cord injury, F-actin rearrangements paralleled by fine structural modifications of the apical domain of ependymal cells were observed. These changes involved disruptions of the apical actin pools as well as fine structural modifications of the microvilli tufts. When comparing the control and injured spinal cords, we also found modifications in the expression of vimentin and glial fibrillary acidic protein (GFAP). After injury, vimentin expression disappeared from the most apical domains of ependymal cells but the number of GFAP-expressing cells within the CC increased. As in other polarized epithelia, the plastic changes in the cytoskeleton may be critically involved in the reaction of ependymal cells following a traumatic injury of the spinal cord.


Subject(s)
Cytoskeleton/metabolism , Ependyma/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord/metabolism , Thoracic Vertebrae/injuries , Animals , Cytoskeleton/pathology , Ependyma/cytology , Ependyma/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spinal Cord/cytology , Spinal Cord/pathology , Spinal Cord Injuries/pathology
15.
Brain Struct Funct ; 226(3): 715-741, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33427974

ABSTRACT

The purinergic system is one of the oldest cell-to-cell communication mechanisms and exhibits relevant functions in the regulation of the central nervous system (CNS) development. Amongst the components of the purinergic system, the ionotropic P2X7 receptor (P2X7R) stands out as a potential regulator of brain pathology and physiology. Thus, P2X7R is known to regulate crucial aspects of neuronal cell biology, including axonal elongation, path-finding, synapse formation and neuroprotection. Moreover, P2X7R modulates neuroinflammation and is posed as a therapeutic target in inflammatory, oncogenic and degenerative disorders. However, the lack of reliable technical and pharmacological approaches to detect this receptor represents a major hurdle in its study. Here, we took advantage of the P2rx7-EGFP reporter mouse, which expresses enhanced green fluorescence protein (EGFP) immediately downstream of the P2rx7 proximal promoter, to conduct a detailed study of its distribution. We performed a comprehensive analysis of the pattern of P2X7R expression in the brain of E18.5 mouse embryos revealing interesting areas within the CNS. Particularly, strong labelling was found in the septum, as well as along the entire neural roof plate zone of the brain, except chorioidal roof areas, but including specialized circumventricular roof formations, such as the subfornical and subcommissural organs (SFO; SCO). Moreover, our results reveal what seems a novel circumventricular organ, named by us postarcuate organ (PArcO). Furthermore, this study sheds light on the ongoing debate regarding the specific presence of P2X7R in neurons and may be of interest for the elucidation of additional roles of P2X7R in the idiosyncratic histologic development of the CNS and related systemic functions.


Subject(s)
Brain/pathology , Circumventricular Organs/pathology , Ependyma/pathology , Neuroglia/pathology , Animals , Brain/metabolism , Circumventricular Organs/metabolism , Ependyma/metabolism , Green Fluorescent Proteins/metabolism , Mice, Transgenic , Neuroglia/metabolism , Neurons/metabolism , Neurons/pathology , Receptors, Purinergic P2X7/metabolism
16.
Semin Cell Dev Biol ; 112: 61-68, 2021 04.
Article in English | MEDLINE | ID: mdl-32771376

ABSTRACT

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.


Subject(s)
Aging/genetics , Ependyma/metabolism , Lipids/genetics , Neural Stem Cells/metabolism , Aging/pathology , Animals , Cell Proliferation/genetics , Central Nervous System/growth & development , Central Nervous System/metabolism , Central Nervous System/pathology , Ependyma/growth & development , Ependyma/pathology , Humans , Lateral Ventricles/growth & development , Lateral Ventricles/metabolism , Lateral Ventricles/pathology , Mice , Neural Stem Cells/physiology , Neurogenesis/genetics , Neurons/metabolism , Neurons/pathology , Telencephalon/metabolism , Telencephalon/pathology
17.
Int J Surg Pathol ; 29(1): 85-89, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32466706

ABSTRACT

Different cellular constituents of the central nervous system occurring in encephaloceles or neuroglial heterotopias (NGHs) have been reported, but the ependymal morphology has rarely been described in the previous literature, let alone the related histological images. To determine the ependymal morphology in encephaloceles or NGHs, we report a rare case of encephalocele with numerous ependymal components. Radiological examination showed that a 6.2 × 3.1 cm nasal dorsum mass-forming encephalocele in a 24-year-old woman, who had an intracranial connection through a frontal bone defect. This patient underwent a resection of the encephalocele under nasal endoscopy and a reconstruction of the cranial base. The patient had a good prognosis with no postoperative complications during follow-up. Microscopically, the ependymal components entrapped in a collagenized background showed numerous slit-like spaces lined by columnar cells with abundant palely eosinophilic cytoplasm and apical surface microvilli. With immunohistochemistry, in addition to the expression of EMA along with the slit-like spaces, GFAP and S100 were diffusely expressed in the slit-like spaces. In conclusion, the ependymal component in either encephaloceles or NGHs may present slit-like spaces arranged in an anastomosing pattern. The unusual morphology of ependyma continues to be underrecognized by pathologists and is easily misdiagnosed; therefore, an awareness of the morphological change in ependyma is necessary.


Subject(s)
Encephalocele/diagnosis , Ependyma/pathology , Frontal Bone/abnormalities , Biomarkers, Tumor/analysis , Diagnosis, Differential , Encephalocele/pathology , Encephalocele/surgery , Endoscopy , Ependyma/surgery , Female , Frontal Bone/diagnostic imaging , Frontal Bone/surgery , Hemangiosarcoma/diagnosis , Humans , Magnetic Resonance Imaging , Tomography, X-Ray Computed , Young Adult
18.
Sci Rep ; 10(1): 12435, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32709945

ABSTRACT

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.


Subject(s)
Cilia/pathology , Cytoskeletal Proteins/genetics , Ependyma/growth & development , Hydrocephalus/genetics , Animals , Animals, Newborn , Axoneme/ultrastructure , Cilia/metabolism , Cilia/ultrastructure , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Ependyma/cytology , Ependyma/pathology , Ependyma/ultrastructure , Female , Humans , Hydrocephalus/congenital , Hydrocephalus/pathology , Intravital Microscopy , Male , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Microscopy, Video
19.
JCI Insight ; 5(9)2020 05 07.
Article in English | MEDLINE | ID: mdl-32229724

ABSTRACT

Hydrocephalus is characterized by abnormal accumulation of cerebrospinal fluid (CSF) in the ventricular cavity. The circulation of CSF in brain ventricles is controlled by the coordinated beating of motile cilia at the surface of ependymal cells (ECs). Here, we show that MT1-MMP is highly expressed in olfactory bulb, rostral migratory stream, and the ventricular system. Mice deficient for membrane-type 1-MMP (MT1-MMP) developed typical phenotypes observed in hydrocephalus, such as dome-shaped skulls, dilated ventricles, corpus callosum agenesis, and astrocyte hypertrophy, during the first 2 weeks of postnatal development. MT1-MMP-deficient mice exhibited reduced and disorganized motile cilia with the impaired maturation of ECs, leading to abnormal CSF flow. Consistent with the defects in motile cilia morphogenesis, the expression of promulticiliogenic genes was significantly decreased, with a concomitant hyperactivation of Notch signaling in the walls of lateral ventricles in Mmp14-/- brains. Inhibition of Notch signaling by γ-secretase inhibitor restored ciliogenesis in Mmp14-/- ECs. Taken together, these data suggest that MT1-MMP is required for ciliogenesis and EC maturation through suppression of Notch signaling during early brain development. Our findings indicate that MT1-MMP is critical for early brain development and loss of MT1-MMP activity gives rise to hydrocephalus.


Subject(s)
Cilia/pathology , Ependyma , Hydrocephalus , Lateral Ventricles , Matrix Metalloproteinase 14/physiology , Animals , Animals, Newborn , Cell Differentiation , Cells, Cultured , Ependyma/metabolism , Ependyma/pathology , Female , Hydrocephalus/metabolism , Hydrocephalus/pathology , Lateral Ventricles/metabolism , Lateral Ventricles/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
Nat Commun ; 11(1): 1860, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32312952

ABSTRACT

Ependymal cells (ECs) are multiciliated neuroepithelial cells that line the ventricles of the brain and the central canal of the spinal cord (SC). How ependymal motile cilia are maintained remains largely unexplored. Here we show that zebrafish embryos deficient in Wnt signaling have defective motile cilia, yet harbor intact basal bodies. With respect to maintenance of ependymal motile cilia, plcδ3a is a target gene of Wnt signaling. Lack of Connexin43 (Cx43), especially its channel function, decreases motile cilia and intercellular Ca2+ wave (ICW) propagation. Genetic ablation of cx43 in zebrafish and mice diminished motile cilia. Finally, Cx43 is also expressed in ECs of the human SC. Taken together, our findings indicate that gap junction mediated ICWs play an important role in the maintenance of ependymal motile cilia, and suggest that the enhancement of functional gap junctions by pharmacological or genetic manipulations may be adopted to ameliorate motile ciliopathy.


Subject(s)
Cilia/metabolism , Connexin 43/metabolism , Connexins/metabolism , Ependyma/metabolism , Spinal Cord/metabolism , Zebrafish/embryology , Animals , Cell Differentiation , Cilia/genetics , Connexin 43/genetics , Ependyma/pathology , Gap Junctions , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Humans , Male , Mice , Mice, Knockout , Signal Transduction/genetics , Wnt Signaling Pathway/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...