Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 371
Filter
1.
Biochem Biophys Res Commun ; 524(2): 497-501, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32008747

ABSTRACT

Circadian rhythms are a fundamental biological phenomena that control various physiological functions. The suprachiasmatic nucleus (SCN) is a master clock that integrates various peripheral clocks. Recently, the choroid plexus (CP) was reported to be one such peripheral clock, a circadian oscillator that might conversely affect the SCN. Hence, the principle aim of our study was to unravel the circadian oscillator within the CP. Quantitative PCR against rPer1, rPer2, and rBmal1 showed that CP in the lateral ventricle (CP-LV) and fourth ventricle (CP-4V) has a robust circadian oscillator. The phases of the CP oscillator are between those of the pineal gland (PG) and SCN. Bioluminescence monitoring of explants showed that the intrinsic circadian period of CP-LV and CP-4V was approximately 21 h, which is shorter than SCN and PG. It is possible that interaction between oscillators of the CP-LV, CP-4V, PG, and SCN ensures the SCN adopts a stable 24 h rhythm, with each of the regions having an intrinsic oscillator with different phases and periods. In situ hybridization analysis revealed that dusk-to-dawn variation of rPer2 expression was found in epithelial cells of the CP only. Furthermore, the CP circadian oscillator might control cerebrospinal fluid secretion. However, no dusk-to-dawn variation in expression of the water channel, aquaporin 1, was observed. Further investigations are needed to clarify the involvement of circadian rhythm on CP.


Subject(s)
Choroid Plexus/physiology , Circadian Rhythm , Animals , Aquaporin 1/analysis , Aquaporin 1/genetics , CLOCK Proteins/analysis , CLOCK Proteins/genetics , Choroid Plexus/ultrastructure , Gene Expression Regulation , Male , Period Circadian Proteins/analysis , Period Circadian Proteins/genetics , Rats , Rats, Transgenic , Rats, Wistar
2.
Cereb Cortex ; 30(6): 3617-3631, 2020 05 18.
Article in English | MEDLINE | ID: mdl-31912879

ABSTRACT

The choroid plexus (CP) is the predominant supplier of cerebral spinal fluid (CSF) and the site of the blood-CSF barrier and is thus essential for brain development and central nervous system homeostasis. Despite these crucial roles, our understanding of the molecular and cellular processes giving rise to the CPs within the ventricles of the mammalian brain is very rudimentary. Here, we identify WNT5a as an important regulator of CP development, where it acts as a pivotal factor driving CP epithelial morphogenesis in all ventricles. We show that WNT5a is essential for the establishment of a cohesive epithelium in the developing CP. We find that in its absence all CPs are substantially reduced in size and complexity and fail to expand into the ventricles. Severe defects were observed in the epithelial cytoarchitecture of all Wnt5a-/- CPs, exemplified by loss of apicobasally polarized morphology and detachment from the ventricular surface and/or basement membrane. We also present evidence that the WNT5a receptor, RYK, and the RHOA kinase, ROCK, are required for normal CP epithelial morphogenesis. Our study, therefore, reveals important insights into the molecular and cellular mechanisms governing CP development.


Subject(s)
Choroid Plexus/embryology , Epithelial Cells/ultrastructure , Receptor Protein-Tyrosine Kinases/genetics , Wnt-5a Protein/genetics , Amides/pharmacology , Animals , Cell Shape/drug effects , Cell Shape/genetics , Choroid Plexus/cytology , Choroid Plexus/drug effects , Choroid Plexus/ultrastructure , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Injections, Intraventricular , Mice , Microinjections , Microscopy, Electron, Transmission , Morphogenesis/genetics , Pyridines/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Wnt-5a Protein/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
3.
Brain Behav Immun ; 79: 216-227, 2019 07.
Article in English | MEDLINE | ID: mdl-30822467

ABSTRACT

Perinatal infection and inflammation are major risk factors for injury in the developing brain, however, underlying mechanisms are not fully understood. Leukocyte migration to the cerebrospinal fluid (CSF) and brain is a hallmark of many pathologies of the central nervous system including those in neonates. We previously reported that systemic activation of Toll-like receptor (TLR) 2, a major receptor for gram-positive bacteria, by agonist Pam3CSK4 (P3C) resulted in dramatic neutrophil and monocyte infiltration to the CSF and periventricular brain of neonatal mice, an effect that was absent by the TLR4 agonist, LPS. Here we first report that choroid plexus is a route of TLR2-mediated leukocyte infiltration to the CSF by performing flow cytometry and transmission electron microscopy (TEM) of the choroid plexus. Next, we exploited the striking discrepancy between P3C and LPS effects on cell migration to determine the pathways regulating leukocyte trafficking through the choroid plexus. We performed RNA sequencing on the choroid plexus after administration of P3C and LPS to postnatal day 8 mice. A cluster gene analysis revealed a TLR2-specific signature of chemotaxis represented by 80-fold increased expression of the gene Ccl3 and 1000-fold increased expression of the gene Cxcl2. Ingenuity pathway analysis (IPA) revealed TLR2-specific molecular signaling related to cytoskeleton organization (e.g. actin signaling) as well as inositol phospholipids biosynthesis and degradation. This included upregulation of genes such as Rac2 and Micall2. In support of IPA results, ultrastructural analysis by TEM revealed clefting and perforations in the basement membrane of the choroid plexus epithelial cells in P3C-treated mice. In summary, we show that the choroid plexus is a route of TLR2-mediated transmigration of neutrophils and monocytes to the developing brain, and reveal previously unrecognized mechanisms that includes a specific chemotaxis profile as well as pathways regulating cytoskeleton and basement membrane remodeling.


Subject(s)
Choroid Plexus/metabolism , Choroid Plexus/ultrastructure , Toll-Like Receptor 2/genetics , Animals , Animals, Newborn , Brain/metabolism , Cell Movement , Central Nervous System/metabolism , Chemotaxis/genetics , Chemotaxis/physiology , Choroid Plexus/physiology , Cytoskeleton/genetics , Cytoskeleton/physiology , Flow Cytometry/methods , Inflammation/metabolism , Leukocytes/metabolism , Leukocytes/physiology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission/methods , Monocytes/metabolism , Neutrophils/metabolism , Toll-Like Receptor 2/metabolism , Transcriptome
4.
Am J Physiol Cell Physiol ; 316(4): C525-C544, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30576237

ABSTRACT

Choroid plexus epithelial cells (CPECs) secrete cerebrospinal fluid (CSF). They express Na+-K+-ATPase and Na+-K+-2Cl- cotransporter 1 (NKCC1) on their apical membrane, deviating from typical basolateral membrane location in secretory epithelia. Given this peculiarity, the direction of basal net ion fluxes mediated by NKCC1 in CPECs is controversial, and cotransporter function is unclear. Determining the direction of basal NKCC1-mediated fluxes is critical to understanding the function of apical NKCC1. If NKCC1 works in the net efflux mode, it may be directly involved in CSF secretion. Conversely, if NKCC1 works in the net influx mode, it would have an absorptive function, contributing to intracellular Cl- concentration ([Cl-]i) and cell water volume (CWV) maintenance needed for CSF secretion. We resolve this long-standing debate by electron microscopy (EM), live-cell-imaging microscopy (LCIM), and intracellular Na+ and Cl- measurements in single CPECs of NKCC1+/+ and NKCC1-/- mouse. NKCC1-mediated ion and associated water fluxes are tightly linked, thus their direction is inferred by measuring CWV changes. Genetic or pharmacological NKCC1 inactivation produces CPEC shrinkage. EM of NKCC1-/- CPECs in situ shows they are shrunken, forming large dilations of their basolateral extracellular spaces, yet remaining attached by tight junctions. Normarski LCIM shows in vitro CPECs from NKCC1-/- are ~17% smaller than NKCC1+/+. CWV measurements in calcein-loaded CPECs show that bumetanide (10 µM) produces ~16% decrease in CWV in NKCC1+/+ but not in NKCC1-/- CPECs. Our findings suggest that under basal conditions apical NKCC1 is continuously active and works in the net inward flux mode maintaining [Cl-]i and CWV needed for CSF secretion.


Subject(s)
Choroid Plexus/drug effects , Choroid Plexus/physiology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 2/genetics , Solute Carrier Family 12, Member 2/metabolism , Animals , Bumetanide/pharmacology , Cells, Cultured , Choroid Plexus/ultrastructure , Epithelial Cells/ultrastructure , Mice , Mice, 129 Strain , Mice, Inbred C57BL
5.
CNS Neurol Disord Drug Targets ; 17(10): 743-756, 2018.
Article in English | MEDLINE | ID: mdl-30179148

ABSTRACT

BACKGROUND & OBJECTIVE: Regulation of composition, volume and turnover of fluids surrounding the brain and damp cells is vital. These fluids transport all substances required for cells and remove the unwanted materials. This regulation tends to act as barrier to prevent free exchange of materials between the brain and blood. There are specific mechanisms concerned with fluid secretion of the controlled composition of the brain, and others responsible for reabsorption eventually to blood and the extracellular fluid whatever their composition is. The current view assumes that choroidal plexuses secrete the major part of Cerebrospinal Fluid (CSF), while the Blood-Brain Barrier (BBB) has a much less contribution to fluid production, generating Interstitial Fluid (ISF) that drains to CSF. The skull is a rigid box; thereby the sum of volumes occupied by the parenchyma with its ISF, related connective tissue, the vasculature, the meninges and the CSF must be relatively constant according to the Monroe-Kellie dogma. This constitutes a formidable challenge that normal organisms surpass daily. The ISF and CSF provide water and solutes influx and efflux from cells to these targeted fluids in a quite precise way. Microvessels within the parenchyma are sufficiently close to every cell where diffusion areas for solutes are tiny. Despite this, CSF and ISF exhibit very similar compositions, but differ significantly from blood plasma. Many hydrophilic substances are effectively prevented from the entry into the brain via blood, while others like neurotransmitters are extremely hindered from getting out of the brain. Anatomical principle of the barrier and routes of fluid transfer cannot explain the extraordinary accuracy of fluids and substances needed to enter or leave the brain firmly. There is one aspect that has not been deeply analyzed, despite being prevalent in all the above processes, it is considered a part of the CSF and ISF dynamics. This aspect is the energy necessary to propel them properly in time, form, space, quantity and temporality. CONCLUSION: The recent hypothesis based on glucose and ATP as sources of energy presents numerous contradictions and controversies. The discovery of the unsuspected intrinsic ability of melanin to dissociate and reform water molecules, similar to the role of chlorophyll in plants, was confirmed in the study of ISF and CSF biology.


Subject(s)
Biological Transport/physiology , Blood-Brain Barrier/physiology , Brain/physiology , Cerebrospinal Fluid/metabolism , Melanins/metabolism , Water-Electrolyte Balance/physiology , Animals , Brain Edema/cerebrospinal fluid , Brain Edema/metabolism , Choroid Plexus/metabolism , Choroid Plexus/ultrastructure , Homeostasis , Humans , Melanins/chemistry
6.
J Neuroinflammation ; 15(1): 50, 2018 Feb 20.
Article in English | MEDLINE | ID: mdl-29463289

ABSTRACT

BACKGROUND: Echovirus (E) 30 (E-30) meningitis is characterized by neuroinflammation involving immune cell pleocytosis at the protective barriers of the central nervous system (CNS). In this context, infection of the blood-cerebrospinal fluid barrier (BCSFB), which has been demonstrated to be involved in enteroviral CNS pathogenesis, may affect the tight junction (TJ) and adherens junction (AJ) function and morphology. METHODS: We used an in vitro human choroid plexus epithelial (HIBCPP) cell model to investigate the effect of three clinical outbreak strains (13-311, 13-759, and 14-397) isolated in Germany in 2013, and compared them to E-30 Bastianni. Conducting transepithelial electrical resistance (TEER), paracellular dextran flux measurement, quantitative real-time polymerase chain reaction (qPCR), western blot, and immunofluorescence analysis, we investigated TJ and AJ function and morphology as well as strain-specific E-30 infection patterns. Additionally, transmission electron and focused ion beam microscopy electron microscopy (FIB-SEM) was used to evaluate the mode of leukocyte transmigration. Genome sequencing and phylogenetic analyses were performed to discriminate potential genetic differences among the outbreak strains. RESULTS: We observed a significant strain-dependent decrease in TEER with strains E-30 Bastianni and 13-311, whereas paracellular dextran flux was only affected by E-30 Bastianni. Despite strong similarities among the outbreak strains in replication characteristics and particle distribution, strain 13-311 was the only outbreak isolate revealing comparable disruptive effects on TJ (Zonula Occludens (ZO) 1 and occludin) and AJ (E-cadherin) morphology to E-30 Bastianni. Notwithstanding significant junctional alterations upon E-30 infection, we observed both para- and transcellular leukocyte migration across HIBCPP cells. Complete genome sequencing revealed differences between the strains analyzed, but no explicit correlation with the observed strain-dependent effects on HIBCPP cells was possible. CONCLUSION: The findings revealed distinct E-30 strain-specific effects on barrier integrity and junctional morphology. Despite E-30-induced barrier alterations leukocyte trafficking did not exclusively occur via the paracellular route.


Subject(s)
Blood-Brain Barrier/virology , Cerebrospinal Fluid/virology , Choroid Plexus/virology , Disease Outbreaks , Enterovirus B, Human/isolation & purification , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/ultrastructure , Cell Line, Tumor , Cell Survival/physiology , Cells, Cultured , Cerebrospinal Fluid/metabolism , Choroid Plexus/metabolism , Choroid Plexus/ultrastructure , Enterovirus B, Human/metabolism , Humans , Phylogeny , Species Specificity
7.
Am J Physiol Cell Physiol ; 314(5): C519-C533, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29351408

ABSTRACT

The choroid plexus epithelial cells (CPECs) belong to a small group of polarized cells, where the Na+-K+-ATPase is expressed in the luminal membrane. The basic polarity of the cells is, therefore, still debated. We investigated the subcellular distribution of an array of proteins known to play fundamental roles either in establishing and maintaining basic cell polarity or in the polarized delivery and recycling of plasma membrane proteins. Immunofluorescence histochemical analysis was applied to determine the subcellular localization of apical and basolateral membrane determinants. Mass spectrometry analysis of CPECs isolated by fluorescence-activated cell sorting was applied to determine the expression of specific forms of the proteins. CPECs mainly express the cell-adhesive P-cadherin, which is localized to the lateral membranes. Proteins belonging to the Crumbs and partitioning defective (Par) protein complexes were all localized to the luminal membrane domain. Par-1 and the Scribble complex were localized to the basolateral membrane domain. Lethal(2) giant larvae homolog 2 (Lgl2) labeling was preferentially observed in the luminal membrane domain. Phosphatidylinositol 3,4,5-trisphosphate (PIP3) was immunolocalized to the basolateral membrane domain, while phosphatidylinositol 4,5-bisphosphate (PIP2) staining was most prominent in the luminal membrane domain along with the PIP3 phosphatase, Pten. The apical target-SNARE syntaxin-3 and the basolateral target-SNARE syntaxin-4 were both localized to the apical membrane domain in CPECs, which lack cellular expression of the clathrin adaptor protein AP-1B for basolateral protein recycling. In conclusion, the CPECs are conventionally polarized, but express P-cadherin at cell-cell contacts, and Lgl2 and syntaxin-4 in the luminal plasma membrane domain.


Subject(s)
Cell Membrane/metabolism , Cell Polarity , Choroid Plexus/metabolism , Epithelial Cells/metabolism , Intercellular Junctions/metabolism , P-Selectin/metabolism , Qa-SNARE Proteins/metabolism , Animals , Cell Membrane/ultrastructure , Choroid Plexus/ultrastructure , Epithelial Cells/ultrastructure , Intercellular Junctions/ultrastructure , Male , Mice, Inbred C57BL , Multiprotein Complexes/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/metabolism , Proteomics/methods , beta Karyopherins/metabolism
8.
J Alzheimers Dis ; 60(1): 43-56, 2017.
Article in English | MEDLINE | ID: mdl-28777754

ABSTRACT

Genome-wide association studies (GWAS) identified susceptibility loci associated with decreased hippocampal volume, and found hippocampal subfield-specific effects at MSRB3 (methionine sulfoxide reductase-B3). The MSRB3 locus was also linked to increased risk for late onset Alzheimer's disease (AD). In this study, we uncovered novel sites of MsrB3 expression in CA pyramidal layer and arteriolar walls by using automated immunohistochemistry on hippocampal sections from 23 individuals accompanied by neuropathology reports and clinical dementia rating scores. Controls, cognitively intact subjects with no hippocampal neurofibrillary tangles, exhibited MsrB3 signal as distinct but rare puncta in CA1 pyramidal neuronal somata. In CA3, however, MsrB3-immunoreactivity was strongest in the neuropil of the pyramidal layer. These patterns were replicated in rodent hippocampi where ultrastructural and immunohistofluorescence analysis revealed MsrB3 signal associated with synaptic vesicles and colocalized with mossy fiber terminals. In AD subjects, the number of CA1 pyramidal neurons with frequent, rather than rare, MsrB3-immunoreactive somatic puncta increased in comparison to controls. This change in CA1 phenotype correlated with the occurrence of AD pathological hallmarks. Moreover, the intensity of MsrB3 signal in the neuropil of CA3 pyramidal layer correlated with the signal pattern in neurons of CA1 pyramidal layer that was characteristic of cognitively intact individuals. Finally, MsrB3 signal in the arteriolar walls in the hippocampal white matter decreased in AD patients. This characterization of GWAS-implicated MSRB3 protein expression in human hippocampus suggests that patterns of neuronal and vascular MsrB3 protein expression reflect or underlie pathology associated with AD.


Subject(s)
Alzheimer Disease/pathology , Hippocampus/metabolism , Hippocampus/pathology , Methionine Sulfoxide Reductases/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Animals , Choroid Plexus/metabolism , Choroid Plexus/pathology , Choroid Plexus/ultrastructure , Ependyma/metabolism , Ependyma/pathology , Ependyma/ultrastructure , Female , Gene Expression Regulation/physiology , Genome-Wide Association Study , Hippocampus/ultrastructure , Humans , Male , Methionine Sulfoxide Reductases/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Electron , Middle Aged , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Pyramidal Cells/ultrastructure , Rats , Rats, Wistar , Vesicle-Associated Membrane Protein 2/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
9.
Am J Physiol Cell Physiol ; 312(6): C673-C686, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28330845

ABSTRACT

The choroid plexus epithelium is a secretory epithelium par excellence. However, this is perhaps not the most prominent reason for the massive interest in this modest-sized tissue residing inside the brain ventricles. Most likely, the dominant reason for extensive studies of the choroid plexus is the identification of this epithelium as the source of the majority of intraventricular cerebrospinal fluid. This finding has direct relevance for studies of diseases and conditions with deranged central fluid volume or ionic balance. While the concept is supported by the vast majority of the literature, the implication of the choroid plexus in secretion of the cerebrospinal fluid was recently challenged once again. Three newer and promising areas of current choroid plexus-related investigations are as follows: 1) the choroid plexus epithelium as the source of mediators necessary for central nervous system development, 2) the choroid plexus as a route for microorganisms and immune cells into the central nervous system, and 3) the choroid plexus as a potential route for drug delivery into the central nervous system, bypassing the blood-brain barrier. Thus, the purpose of this review is to highlight current active areas of research in the choroid plexus physiology and a few matters of continuous controversy.


Subject(s)
Cerebrospinal Fluid/physiology , Choroid Plexus/physiology , Epithelium/physiology , Ion Channels/metabolism , Signal Transduction/physiology , Animals , Biological Transport , Blood-Brain Barrier , Choroid Plexus/ultrastructure , Gene Expression , Humans , Hydrogen-Ion Concentration , Intercellular Adhesion Molecule-1/cerebrospinal fluid , Intercellular Adhesion Molecule-1/genetics , Ion Channels/antagonists & inhibitors , Ion Channels/genetics , Membrane Transport Modulators/pharmacology , Vascular Cell Adhesion Molecule-1/cerebrospinal fluid , Vascular Cell Adhesion Molecule-1/genetics
10.
Physiol Rep ; 5(1)2017 Jan.
Article in English | MEDLINE | ID: mdl-28053225

ABSTRACT

The cerebrospinal fluid (CSF) pH influences brain interstitial pH and, therefore, brain function. We hypothesized that the choroid plexus epithelium (CPE) expresses the vacuolar H+-ATPase (V-ATPase) as an acid extrusion mechanism in the luminal membrane to counteract detrimental elevations in CSF pH. The expression of mRNA corresponding to several V-ATPase subunits was demonstrated by RT-PCR analysis of CPE cells (CPECs) isolated by fluorescence-activated cell sorting. Immunofluorescence and electron microscopy localized the V-ATPase primarily in intracellular vesicles with only a minor fraction in the luminal microvillus area. The vesicles did not translocate to the luminal membrane in two in vivo models of hypocapnia-induced alkalosis. The Na+-independent intracellular pH (pHi) recovery from acidification was studied in freshly isolated clusters of CPECs. At extracellular pH (pHo) 7.4, the cells failed to display significant concanamycin A-sensitive pHi recovery (i.e., V-ATPase activity). The recovery rate in the absence of Na+ amounted to <10% of the pHi recovery rate observed in the presence of Na+ Recovery of pHi was faster at pHo 7.8 and was abolished at pHo 7.0. The concanamycin A-sensitive pHi recovery was stimulated by cAMP at pH 7.4 in vitro, but intraventricular infusion of the membrane-permeant cAMP analog 8-CPT-cAMP did not result in trafficking of the V-ATPase. In conclusion, we find evidence for the expression of a minor fraction of V-ATPase in the luminal membrane of CPECs. This fraction does not contribute to enhanced acid extrusion at high extracellular pH, but seems to be activated by cAMP in a trafficking-independent manner.


Subject(s)
Cell Membrane/chemistry , Choroid Plexus/metabolism , Hydrogen-Ion Concentration/drug effects , Intracellular Fluid/chemistry , Vacuolar Proton-Translocating ATPases/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/administration & dosage , 8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate/metabolism , Animals , Brain/physiology , Cell Membrane/metabolism , Cerebrospinal Fluid/chemistry , Cerebrospinal Fluid/enzymology , Cerebrospinal Fluid/physiology , Choroid Plexus/chemistry , Choroid Plexus/cytology , Choroid Plexus/ultrastructure , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Flow Cytometry , Intracellular Fluid/enzymology , Intracellular Fluid/metabolism , Macrolides/administration & dosage , Macrolides/adverse effects , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Sodium/metabolism , Thionucleotides/metabolism
11.
Adv Exp Med Biol ; 913: 397-402, 2016.
Article in English | MEDLINE | ID: mdl-27796901

ABSTRACT

It is well proved already that neurogenesis does take place in mammals' brain, including human brain. However, neurogenesis by itself is not able to compensate for brain tissue loss in serious neurological diseases, such as stroke, brain trauma or neurodegenerative disorders. Recent evidences show that neural stem cell niches are present not only in classical locations, such as subventricularor subgranular zones, but in other areas as well, including tissues contiguous to the brain (meninges and choroid plexus).In this chapter we revise the relationship of neural stem cells with interstitial cells (mainly telocytes), which we think is significant, and we describe what is known about the juxtacerebral tissue neurogenesis potential.


Subject(s)
Choroid Plexus/physiology , Meninges/physiology , Nerve Regeneration/physiology , Neural Stem Cells/physiology , Stem Cell Niche/physiology , Telocytes/physiology , Animals , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/rehabilitation , Choroid Plexus/ultrastructure , Hippocampus/physiology , Hippocampus/ultrastructure , Humans , Lateral Ventricles/physiology , Lateral Ventricles/ultrastructure , Meninges/ultrastructure , Microscopy, Electron, Transmission , Neural Stem Cells/ultrastructure , Neurogenesis/physiology , Rats , Stroke/pathology , Stroke Rehabilitation , Telocytes/ultrastructure
12.
Nat Commun ; 7: 11876, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27336173

ABSTRACT

Maintenance of epithelial cell polarity and epithelial barrier relies on the spatial organization of the actin cytoskeleton and proper positioning/assembly of intercellular junctions. However, how these processes are regulated is poorly understood. Here we reveal a key role for the multifunctional protein Alix in both processes. In a knockout mouse model of Alix, we identified overt structural changes in the epithelium of the choroid plexus and in the ependyma, such as asymmetrical cell shape and size, misplacement and abnormal beating of cilia, blebbing of the microvilli. These defects culminate in excessive cell extrusion, enlargement of the lateral ventricles and hydrocephalus. Mechanistically, we find that by interacting with F-actin, the Par complex and ZO-1, Alix ensures the formation and maintenance of the apically restricted actomyosin-tight junction complex. We propose that in this capacity Alix plays a role in the establishment of apical-basal polarity and in the maintenance of the epithelial barrier.


Subject(s)
Actomyosin/metabolism , Blood-Brain Barrier , Calcium-Binding Proteins/physiology , Choroid Plexus/metabolism , Tight Junctions/metabolism , Actins/metabolism , Animals , Cell Polarity , Choroid Plexus/ultrastructure , Ependyma/ultrastructure , Epithelial Cells/ultrastructure , Hydrocephalus/etiology , Mice , Mice, Knockout , Zonula Occludens-1 Protein/metabolism
13.
Nat Cell Biol ; 18(4): 418-30, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26999738

ABSTRACT

Aberrant Notch signalling has been linked to many cancers including choroid plexus (CP) tumours, a group of rare and predominantly paediatric brain neoplasms. We developed animal models of CP tumours, by inducing sustained expression of Notch1, that recapitulate properties of human CP tumours with aberrant NOTCH signalling. Whole-transcriptome and functional analyses showed that tumour cell proliferation is associated with Sonic Hedgehog (Shh) in the tumour microenvironment. Unlike CP epithelial cells, which have multiple primary cilia, tumour cells possess a solitary primary cilium as a result of Notch-mediated suppression of multiciliate differentiation. A Shh-driven signalling cascade in the primary cilium occurs in tumour cells but not in epithelial cells. Lineage studies show that CP tumours arise from monociliated progenitors in the roof plate characterized by elevated Notch signalling. Abnormal SHH signalling and distinct ciliogenesis are detected in human CP tumours, suggesting the SHH pathway and cilia differentiation as potential therapeutic avenues.


Subject(s)
Cell Proliferation/genetics , Choroid Plexus Neoplasms/genetics , Hedgehog Proteins/genetics , Receptor, Notch1/genetics , Animals , Blotting, Western , Choroid Plexus/metabolism , Choroid Plexus/pathology , Choroid Plexus/ultrastructure , Choroid Plexus Neoplasms/metabolism , Choroid Plexus Neoplasms/pathology , Cilia/metabolism , Cilia/ultrastructure , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Humans , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis , Receptor, Notch1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Tumor Cells, Cultured , Tumor Microenvironment/genetics
14.
J Vis Exp ; (101): e52991, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-26273731

ABSTRACT

The choroid plexus is located in the ventricular wall of the brain, the main function of which is believed to be production of cerebrospinal fluid. Choroid plexus epithelial cells (CPECs) covering the surface of choroid plexus tissue harbor multiple unique cilia, but most of the functions of these cilia remain to be investigated. To uncover the function of CPEC cilia with particular reference to their motility, an ex vivo observation system was developed to monitor ciliary motility during embryonic, perinatal and postnatal periods. The choroid plexus was dissected out of the brain ventricle and observed under a video-enhanced contrast microscope equipped with differential interference contrast optics. Under this condition, a simple and quantitative method was developed to analyze the motile profiles of CPEC cilia for several hours ex vivo. Next, the morphological changes of cilia during development were observed by scanning electron microscopy to elucidate the relationship between the morphological maturity of cilia and motility. Interestingly, this method could delineate changes in the number and length of cilia, which peaked at postnatal day (P) 2, while the beating frequency reached a maximum at P10, followed by abrupt cessation at P14. These techniques will enable elucidation of the functions of cilia in various tissues. While related techniques have been published in a previous report(1), the current study focuses on detailed techniques to observe the motility and morphology of CPEC cilia ex vivo.


Subject(s)
Cell Movement/physiology , Choroid Plexus/cytology , Microscopy, Electron, Scanning/methods , Animals , Choroid Plexus/ultrastructure , Cilia/physiology , Cilia/ultrastructure , Computer Systems , Epithelial Cells/cytology , Epithelial Cells/ultrastructure , Image Processing, Computer-Assisted , Mice , Microscopy, Electron, Scanning/instrumentation
15.
Eur. j. anat ; 19(2): 125-129, abr. 2015. ilus, graf
Article in English | IBECS | ID: ibc-141201

ABSTRACT

Hydrocephalus is an accumulation of cerebrospinal fluid (CSF) with dilatation of brain ventricles which can be either communicating or non-communicating. Multiple pathophysiological mechanisms underlie the appearance of hydrocephalus, which has many different causes including birth defects, brain hemorrhage, infection, meningitis, tumor, or head injury. The choroid plexuses (ChP) are circumventricular structures closely related to the above-mentioned pathophysiological mechanisms of the CSF, and aquaporin-1 (AQP1) is the water channel directly implicated in CSF production. Our studies with hydrocephalic rats revealed an increase and redistribution of AQP1 in the ChP, with AQP1 being expressed not only in the cell apical pole, but also in the cell basal pole and in the stroma. The immunohistochemical changes observed in both communicating and non-communicating hydrocephalus suggest a variation in the efficiency of the cells of the ChP, where AQP1 could perform both CSF production and reabsorption in order to delay ventricular dilatation


No disponible


Subject(s)
Animals , Rats , Aquaporin 1 , Hydrocephalus/physiopathology , Choroid Plexus/ultrastructure , Disease Models, Animal
16.
Nat Commun ; 4: 2123, 2013.
Article in English | MEDLINE | ID: mdl-23828504

ABSTRACT

Loss of folate receptor-α function is associated with cerebral folate transport deficiency and childhood-onset neurodegeneration. To clarify the mechanism of cerebral folate transport at the blood-cerebrospinal fluid barrier, we investigate the transport of 5-methyltetrahydrofolate in polarized cells. Here we identify folate receptor-α-positive intralumenal vesicles within multivesicular bodies and demonstrate the directional cotransport of human folate receptor-α, and labelled folate from the basolateral to the apical membrane in rat choroid plexus cells. Both the apical medium of folate receptor-α-transfected rat choroid plexus cells and human cerebrospinal fluid contain folate receptor-α-positive exosomes. Loss of folate receptor-α-expressing cerebrospinal fluid exosomes correlates with severely reduced 5-methyltetrahydrofolate concentration, corroborating the importance of the folate receptor-α-mediated folate transport in the cerebrospinal fluid. Intraventricular injections of folate receptor-α-positive and -negative exosomes into mouse brains demonstrate folate receptor-α-dependent delivery of exosomes into the brain parenchyma. Our results unravel a new pathway of folate receptor-α-dependent exosome-mediated folate delivery into the brain parenchyma and opens new avenues for cerebral drug targeting.


Subject(s)
Choroid Plexus/cytology , Choroid Plexus/metabolism , Exosomes/metabolism , Folic Acid/metabolism , Transcytosis , Adolescent , Adult , Animals , Cell Polarity/drug effects , Child , Choroid Plexus/ultrastructure , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/metabolism , Dogs , Exosomes/drug effects , Exosomes/ultrastructure , Female , Folate Receptor 1/metabolism , Humans , Madin Darby Canine Kidney Cells , Male , Mice , Models, Biological , Monensin/pharmacology , Protein Transport/drug effects , Proton-Coupled Folate Transporter/metabolism , Rats , Tetrahydrofolates/metabolism , Transcytosis/drug effects , Transferrin/pharmacology , Young Adult
17.
Microsc Res Tech ; 75(8): 1104-12, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22434550

ABSTRACT

Mammalian choroid plexuses (CPs) are vascularized structures involved in numerous exchange processes that supply nutrients and hormones to the brain, and that remove deleterious compounds and metabolites from the brain. Studies in the adult Mediterranean buffalo have investigated the morphology of CPs using histochemical and immunohistochemical techniques. To date, however, there have been no studies conducted on ruminants regarding this removal process which serves to repair functional vascular damage in the CPs. Each of these vascular repair processes is a very complex and none of these has not yet been completely understood. Then, the aim of the present study is to investigate the morphological processes during angiogenesis in the CPs of healthy adult buffaloes, utilizing transmission electron microscopy (TEM), scanning electron microscopy (SEM), and immunogold-labeling SEM analysis (biomarkers: angiopoietin-2 [Ang-2], vascular endothelial growth factor receptor-3 [VEGFR-3], and CD133). At TEM, the inner surface of the blood capillaries sometimes showed pillar-like cells, which in contact with endothelial cells formed prominences, which in turn formed neo-blood capillaries. With immunogold-labeling SEM analysis, the CP blood capillaries showed Ang-2 and VEGF-3, respectively, in positive particles and spheroid formations. In addition, the external surface of the blood capillaries showed spheroid formations that originated from the neo-vascular capillaries whose terminals formed a capillary network, positive to CD133. On the basis of these results, the following hypothesis can be made, namely, that these CPs are vascular structures which play a fundamental role in maintaining brain homeostasis and self-repairing of functional vascular damage, independently of the presence of rete mirabile in this species.


Subject(s)
Buffaloes/physiology , Choroid Plexus/anatomy & histology , Immunohistochemistry , Neovascularization, Physiologic , AC133 Antigen , Angiopoietin-2/analysis , Animals , Antigens, CD/analysis , Biomarkers/analysis , Buffaloes/anatomy & histology , Capillaries/physiology , Choroid Plexus/chemistry , Choroid Plexus/physiology , Choroid Plexus/ultrastructure , Glycoproteins/analysis , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Peptides/analysis , Species Specificity , Vascular Endothelial Growth Factor Receptor-3/analysis
18.
J Cereb Blood Flow Metab ; 32(1): 93-104, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21829211

ABSTRACT

The invasion of inflammatory cells occurring after ischemic or traumatic brain injury (TBI) has a detrimental effect on neuronal survival and functional recovery after injury. We have recently demonstrated that not only the blood-brain barrier, but also the blood-cerebrospinal fluid (CSF) barrier (BCSFB), has a role in posttraumatic recruitment of neutrophils. Here, we show that TBI results in a rapid increase in synthesis and release into the CSF of a major chemoattractant for monocytes, CCL2, by the choroid plexus epithelium, a site of the BCSFB. Using an in vitro model of the BCSFB, we also show that CCL2 is released across the apical and basolateral membranes of the choroidal epithelium, a pattern of chemokine secretion that promotes leukocyte migration across epithelial barriers. Immunohistochemical and electron microscopic analyses of choroidal tissue provide evidence for the movement of monocytes, sometimes in tandem with neutrophils, along the paracellular pathways between adjacent epithelial cells. These data further support the pathophysiological role of BCSFB in promoting the recruitment of inflammatory cells to the injured brain.


Subject(s)
Blood-Brain Barrier/immunology , Brain Injuries/immunology , Cerebrospinal Fluid/immunology , Choroid Plexus/immunology , Monocytes/cytology , Animals , Basement Membrane/immunology , Basement Membrane/ultrastructure , Blood-Brain Barrier/ultrastructure , Blotting, Western , Brain Injuries/blood , Brain Injuries/cerebrospinal fluid , Cells, Cultured , Cerebrospinal Fluid/cytology , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Chemotaxis, Leukocyte/immunology , Choroid Plexus/blood supply , Choroid Plexus/ultrastructure , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/immunology , Intercellular Junctions/immunology , Intercellular Junctions/ultrastructure , Male , Microscopy, Electron, Transmission , Monocytes/immunology , Monocytes/ultrastructure , Neutrophil Infiltration/immunology , Rats , Rats, Long-Evans , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
19.
Ann Anat ; 193(3): 197-204, 2011 May.
Article in English | MEDLINE | ID: mdl-21466950

ABSTRACT

Environmental scanning electron microscopy (ESEM) allows the examination of hydrated and dried specimens without a conductive metal coating which could be advantageous in the imaging of biological and medical objects. The aim of this study was to assess the performance and benefits of wet-mode and low vacuum ESEM in comparison to high vacuum scanning electron microscopy (SEM) using the choroid plexus of chicken embryos as a model, an organ of the brain involved in the formation of cerebrospinal fluid in vertebrates. Specimens were fixed with or without heavy metals and examined directly or after critical point drying with or without metal coating. For wet mode ESEM freshly excised specimens without any pre-treatment were also examined. Conventional high vacuum SEM revealed the characteristic morphology of the choroid plexus cells at a high resolution and served as reference. With low vacuum ESEM of dried but uncoated samples the structure appeared well preserved but charging was a problem. It could be reduced by a short beam dwell time and averaging of images or by using the backscattered electron detector instead of the gaseous secondary electron detector. However, resolution was lower than with conventional SEM. Wet mode imaging was only possible with tissue that had been stabilized by fixation. Not all surface details (e.g. microvilli) could be visualized and other structures, like the cilia, were deformed. In summary, ESEM is an additional option for the imaging of bio-medical samples but it is problematic with regard to resolution and sample stability during imaging.


Subject(s)
Choroid Plexus/ultrastructure , Microscopy, Electron, Scanning/instrumentation , Microscopy, Electron, Scanning/methods , Animals , Chick Embryo , Reproducibility of Results , Sensitivity and Specificity , Surface Properties , Vacuum
20.
Microsc Res Tech ; 74(1): 67-75, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21181712

ABSTRACT

The choroid plexuses (CPs) in mammals produce the cerebrospinal fluid (CSF). In the literature, the morphology of CPs and the process that regulates the production of CSF are virtually nonexistent for domestic ruminants. Thus this study has two aims: 1. to investigate the morpho-structure of the buffalo CP microvasculature utilizing light microscopy (LM), scanning electron microscopy (SEM) and transmission electron microscopy (TEM) techniques, and 2. to investigate the relationship between the blood vessels and both the elongated cells and the cells with multiple protrusions located in the CPs. SEM and TEM analyses of the CPs from buffalo brain showed morphological and structural features similar those reported in other mammalian species. Moreover the blood microvasculature is the major component responsible for the formation of the CSF, secreted by the encephalic CPs. In addition the chemical composition of this fluid depends on several morpho-functional characteristics of the vascularization of the CPs. These characteristics are as follows: two shapes of the vascular organization: lamina-like and ovoid-like elongated cells of the CPs, which connect the ventricular cavities to the blood capillaries; and the CP capillaries have diverse forms. In the present study the employment of NADPHd and NOS I was taken as indirect evidence for the presence of NO for investigation their specific role in CPs. Then NOS I immunoreactivity is found in the walls of CP blood vessels demonstrating indirectly the presence of NO with a vaso-dilatatory and autoregulation function of vascular tone by cholinergic nerve stimulation of blood vessel smooth muscle.


Subject(s)
Choroid Plexus/anatomy & histology , Choroid Plexus/blood supply , Microvessels/anatomy & histology , Animals , Buffaloes , Choroid Plexus/metabolism , Choroid Plexus/ultrastructure , Histocytochemistry , Immunohistochemistry , Microscopy , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Microvessels/metabolism , Microvessels/ultrastructure , Nitric Oxide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...