Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
Cereb Cortex ; 34(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38912605

ABSTRACT

Glymphatic dysfunction has been correlated with cognitive decline, with a higher choroid plexus volume (CPV) being linked to a slower glymphatic clearance rate. Nevertheless, the interplay between CPV, glymphatic function, and cognitive impairment in white matter hyperintensities (WMHs) has not yet been investigated. In this study, we performed neuropsychological assessment, T1-weighted three-dimensional (3D-T1) images, and diffusion tensor imaging (DTI) in a cohort of 206 WMHs subjects and 43 healthy controls (HCs) to further explore the relationship. The DTI analysis along the perivascular space (DTI-ALPS) index, as a measure of glymphatic function, was calculated based on DTI. Severe WMHs performed significantly worse in information processing speed (IPS) than other three groups, as well as in executive function than HCs and mild WMHs. Additionally, severe WMHs demonstrated lower DTI-ALPS index and higher CPV than HCs and mild WMHs. Moderate WMHs displayed higher CPV than HCs and mild WMHs. Mini-Mental State Examination, IPS, and executive function correlated negatively with CPV but positively with DTI-ALPS index in WMHs patients. Glymphatic function partially mediated the association between CPV and IPS, indicating a potential mechanism for WMHs-related cognitive impairment. CPV may act as a valuable prognostic marker and glymphatic system as a promising therapeutic target for WMHs-related cognitive impairment.


Subject(s)
Choroid Plexus , Cognitive Dysfunction , Diffusion Tensor Imaging , Glymphatic System , White Matter , Humans , Male , Female , Choroid Plexus/diagnostic imaging , Choroid Plexus/pathology , Choroid Plexus/physiopathology , White Matter/diagnostic imaging , White Matter/pathology , Aged , Glymphatic System/diagnostic imaging , Glymphatic System/pathology , Glymphatic System/physiopathology , Middle Aged , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/pathology , Neuropsychological Tests , Magnetic Resonance Imaging/methods , Processing Speed
2.
Science ; 374(6566): 439-448, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34672740

ABSTRACT

Up to 40% of patients with inflammatory bowel disease present with psychosocial disturbances. We previously identified a gut vascular barrier that controls the dissemination of bacteria from the intestine to the liver. Here, we describe a vascular barrier in the brain choroid plexus (PVB) that is modulated in response to intestinal inflammation through bacteria-derived lipopolysaccharide. The inflammatory response induces PVB closure after gut vascular barrier opening by the up-regulation of the wingless-type, catenin-beta 1 (Wnt/ß-catenin) signaling pathway, rendering it inaccessible to large molecules. In a model of genetically driven closure of choroid plexus endothelial cells, we observed a deficit in short-term memory and anxiety-like behavior, suggesting that PVB closure may correlate with mental deficits. Inflammatory bowel disease­related mental symptoms may thus be the consequence of a deregulated gut­brain vascular axis.


Subject(s)
Choroid Plexus/blood supply , Choroid Plexus/physiopathology , Colitis, Ulcerative/physiopathology , Colitis, Ulcerative/psychology , Intestines/physiopathology , Memory Disorders/physiopathology , Memory, Short-Term , Animals , Anxiety/etiology , Anxiety/physiopathology , Blood-Brain Barrier/pathology , Colitis, Ulcerative/complications , Dextrans , Disease Models, Animal , Humans , Lipopolysaccharides , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Microglia/pathology , Signal Transduction , Tight Junctions/pathology , Wnt Proteins/metabolism , beta Catenin/metabolism
3.
Nature ; 595(7868): 565-571, 2021 07.
Article in English | MEDLINE | ID: mdl-34153974

ABSTRACT

Although SARS-CoV-2 primarily targets the respiratory system, patients with and survivors of COVID-19 can suffer neurological symptoms1-3. However, an unbiased understanding of the cellular and molecular processes that are affected in the brains of patients with COVID-19 is missing. Here we profile 65,309 single-nucleus transcriptomes from 30 frontal cortex and choroid plexus samples across 14 control individuals (including 1 patient with terminal influenza) and 8 patients with COVID-19. Although our systematic analysis yields no molecular traces of SARS-CoV-2 in the brain, we observe broad cellular perturbations indicating that barrier cells of the choroid plexus sense and relay peripheral inflammation into the brain and show that peripheral T cells infiltrate the parenchyma. We discover microglia and astrocyte subpopulations associated with COVID-19 that share features with pathological cell states that have previously been reported in human neurodegenerative disease4-6. Synaptic signalling of upper-layer excitatory neurons-which are evolutionarily expanded in humans7 and linked to cognitive function8-is preferentially affected in COVID-19. Across cell types, perturbations associated with COVID-19 overlap with those found in chronic brain disorders and reside in genetic variants associated with cognition, schizophrenia and depression. Our findings and public dataset provide a molecular framework to understand current observations of COVID-19-related neurological disease, and any such disease that may emerge at a later date.


Subject(s)
Astrocytes/pathology , Brain/pathology , COVID-19/diagnosis , COVID-19/pathology , Choroid Plexus/pathology , Microglia/pathology , Neurons/pathology , Aged , Aged, 80 and over , Brain/metabolism , Brain/physiopathology , Brain/virology , COVID-19/genetics , COVID-19/physiopathology , Cell Nucleus/genetics , Choroid Plexus/metabolism , Choroid Plexus/physiopathology , Choroid Plexus/virology , Female , Humans , Inflammation/virology , Male , Middle Aged , SARS-CoV-2/growth & development , SARS-CoV-2/pathogenicity , Single-Cell Analysis , Transcriptome , Virus Replication
4.
J Cereb Blood Flow Metab ; 41(10): 2699-2711, 2021 10.
Article in English | MEDLINE | ID: mdl-33906512

ABSTRACT

Cerebrospinal fluid (CSF) and interstitial fluid exchange have been shown to increase following pharmacologically-manipulated increases in cerebral arterial pulsatility, consistent with arterial pulsatility improving CSF circulation along perivascular glymphatic pathways. The choroid plexus (CP) complexes produce CSF, and CP activity may provide a centralized indicator of perivascular flow. We tested the primary hypothesis that elevated cortical cerebral blood volume and flow, present in sickle cell disease (SCD), is associated with fractionally-reduced CP perfusion relative to healthy adults, and the supplementary hypothesis that reduced arterial patency, present in moyamoya vasculopathy, is associated with elevated fractional CP perfusion relative to healthy adults. Participants (n = 75) provided informed consent and were scanned using a 3-Tesla arterial-spin-labeling MRI sequence for CP and cerebral gray matter (GM) perfusion quantification. ANOVA was used to calculate differences in CP-to-GM perfusion ratios between groups, and regression analyses applied to evaluate the dependence of the CP-to-GM perfusion ratio on group after co-varying for age and sex. ANOVA yielded significant (p < 0.001) group differences, with CP-to-GM perfusion ratios increasing between SCD (ratio = 0.93 ± 0.28), healthy (ratio = 1.04 ± 0.32), and moyamoya (ratio = 1.29 ± 0.32) participants, which was also consistent with regression analyses. Findings are consistent with CP perfusion being inversely associated with cortical perfusion.


Subject(s)
Anemia, Sickle Cell/physiopathology , Choroid Plexus/physiopathology , Glymphatic System/physiopathology , Moyamoya Disease/physiopathology , Vascular Diseases/physiopathology , Adult , Female , Humans , Male
5.
Ophthalmic Genet ; 42(3): 344-348, 2021 06.
Article in English | MEDLINE | ID: mdl-33641574

ABSTRACT

Background: Norrie disease is a genetic disorder of the retina characterized by impaired retinal vascular development leading to retinal detachment and blindness. Non-retinal manifestations of the disorder include intellectual disability and seizure disorders. However, to date, no association with neurological mass lesions has been described.Materials and methods: Case reporResults: Here, we report a case of a patient with Norrie disease who presented with an enhancing mass of the choroid plexus that spontaneously diminished in size. Conclusion: This report suggests watchful waiting as a reasonable clinical approach to choroid plexus lesions in patients with Norrie disease.


Subject(s)
Blindness/congenital , Brain Diseases/diagnostic imaging , Choroid Plexus/diagnostic imaging , Eye Proteins/genetics , Genetic Diseases, X-Linked/diagnosis , Mutation/genetics , Nerve Tissue Proteins/genetics , Nervous System Diseases/diagnosis , Retinal Degeneration/diagnosis , Spasms, Infantile/diagnosis , Blindness/diagnosis , Blindness/genetics , Brain Diseases/physiopathology , Choroid Plexus/physiopathology , Genetic Diseases, X-Linked/genetics , Gestational Age , Humans , Infant , Magnetic Resonance Imaging , Male , Nervous System Diseases/genetics , Retinal Degeneration/genetics , Spasms, Infantile/genetics
6.
Int Rev Neurobiol ; 154: 413-436, 2020.
Article in English | MEDLINE | ID: mdl-32739013

ABSTRACT

The cerebral vasculature serves as the crossroads of the CNS, supporting exchange of nutrients, metabolic wastes, solutes and cells between the compartments of the brain, including the blood, brain interstitium, and cerebrospinal fluid (CSF). The blood-brain barrier (BBB) regulates the entry and efflux of molecules into brain tissue. The cells of the neurovascular unit regulate cerebral blood flow, matching local metabolic demand to blood supply. The blood-CSF barrier at the choroid plexus secretes CSF, which supports the brain and provides a sink for interstitial solutes not cleared across the BBB. Recent studies have characterized the glymphatic system, a brain-wide network of perivascular spaces that supports CSF and interstitial fluid exchange and the clearance of interstitial solutes to the CSF. The critical role that these structures play in maintaining brain homeostasis is illustrated by the established and emerging roles that their dysfunctions play in the development of neurodegenerative diseases, such as Alzheimer's disease (AD). Loss of BBB and blood-CSF barrier function is reported both in rodent models of AD, and in human AD subjects. Cerebrovascular dysfunction and ischemic injury are well established contributors to both vascular dementia and to a large proportion of cases of sporadic AD. In animal models, the slowed glymphatic clearance of interstitial proteins, such as amyloid ß or tau, are proposed to contribute to the development of neurodegenerative diseases, including AD. In total, these findings suggest that cellular and molecular changes occurring within and around the cerebral vasculature are among the key drivers of neurodegenerative disease pathogenesis.


Subject(s)
Aging , Blood-Brain Barrier , Cerebrospinal Fluid , Cerebrovascular Circulation , Choroid Plexus , Diabetes Mellitus, Type 2 , Glymphatic System , Neurodegenerative Diseases , Aging/metabolism , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Cerebrospinal Fluid/metabolism , Cerebrovascular Circulation/physiology , Choroid Plexus/metabolism , Choroid Plexus/physiopathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Glymphatic System/metabolism , Glymphatic System/physiopathology , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology
7.
Neurobiol Aging ; 89: 108-117, 2020 05.
Article in English | MEDLINE | ID: mdl-32107064

ABSTRACT

The choroid plexus (ChP) is a major source of cerebrospinal fluid (CSF) production, with a direct and indirect role in protein clearance, and pathogenesis of Alzheimer's disease (AD). Here, we tested the link between the ChP volume and levels of CSF proteins in 2 data sets of (i) healthy controls, mild cognitive impairment (MCI), and AD patients from the Alzheimer's Disease Neuroimaging Initiative (ADNI) (N = 509), and (ii) healthy controls and Parkinson's disease (PD) patients from the Parkinson's Progression Markers Initiative (N = 302). All patients had baseline CSF proteins (amyloid-ß, total and phosphorylated-tau and α-synuclein (only in Parkinson's Progression Markers Initiative)). ChP was automatically segmented on 3T structural T1-weighted MRIs. We found negative associations between ChP volume and CSF proteins, which were stronger in healthy controls, early-MCI patients, and PD patients compared with late-MCI and AD patients. Further grouping of patients of ADNI dataset into amyloid-positive and amyloid-negative based on their florbetapir (AV45) PET imaging showed that the association between ChP volume and CSF proteins (t/p-tau) was lower in amyloid-positive group. Our findings support the possible role of ChP in the clearance of CSF proteins, provide evidence for ChP dysfunction in AD, and suggest the need to account for the ChP volume in future studies of CSF-based biomarkers.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/pathology , Amyloid beta-Peptides/cerebrospinal fluid , Choroid Plexus/pathology , Choroid Plexus/physiopathology , Organ Size , Parkinson Disease/diagnosis , Parkinson Disease/pathology , alpha-Synuclein/cerebrospinal fluid , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/physiopathology , Biomarkers/cerebrospinal fluid , Choroid Plexus/diagnostic imaging , Parkinson Disease/diagnostic imaging , Parkinson Disease/physiopathology , Positron-Emission Tomography , tau Proteins
8.
J Neurosci Methods ; 329: 108478, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31669338

ABSTRACT

Acute lymphoblastic leukaemia represents the most common paediatric malignancy. Although survival rates approach up to 90% in children, investigation of leukaemic infiltration into the central nervous system (CNS) is essential due to the presence of ongoing fatal complications. Recent in vitro studies mostly employed models of the blood-brain barrier (BBB), as endothelial cells of the microvasculature represent the largest surface between the blood stream and the brain parenchyma. However, crossing the blood-cerebrospinal fluid barrier (BCSFB) within the choroid plexus (CP) has been shown to be a general capability of leukaemic blasts. Hence, in vitro models of the BCSFB to study leukaemic transmigration may be of major importance to understand the development of CNS leukaemia. This review will summarise available in vitro models of the BCSFB employed to study the cellular interactions with leukaemic blasts during cancer cell transmigration into the brain compartment across primary or immortal/immortalised BCSFB cells. It will also provide an outlook on prospective improvements in BCSFB in vitro models by developing barrier-on-a-chip models and brain organoids.


Subject(s)
Blood-Brain Barrier/physiology , Cell Line, Tumor , Cerebrospinal Fluid/physiology , Choroid Plexus/physiopathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Primary Cell Culture , Transcellular Cell Migration/physiology , Animals , Humans
9.
Genes Dev ; 33(23-24): 1635-1640, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31624084

ABSTRACT

Short tandem repeats (STRs) are prone to expansion mutations that cause multiple hereditary neurological and neuromuscular diseases. To study pathomechanisms using mouse models that recapitulate the tissue specificity and developmental timing of an STR expansion gene, we used rolling circle amplification and CRISPR/Cas9-mediated genome editing to generate Dmpk CTG expansion (CTGexp) knockin models of myotonic dystrophy type 1 (DM1). We demonstrate that skeletal muscle myoblasts and brain choroid plexus epithelial cells are particularly susceptible to Dmpk CTGexp mutations and RNA missplicing. Our results implicate dysregulation of muscle regeneration and cerebrospinal fluid homeostasis as early pathogenic events in DM1.


Subject(s)
Alternative Splicing/genetics , Microsatellite Repeats/genetics , Muscle, Skeletal/physiopathology , Myotonic Dystrophy/genetics , Myotonic Dystrophy/physiopathology , RNA Splicing/genetics , 3' Untranslated Regions/genetics , Animals , Choroid Plexus/physiopathology , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/cytology , Mutation , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , RNA-Binding Proteins/genetics
10.
JCI Insight ; 4(11)2019 06 06.
Article in English | MEDLINE | ID: mdl-31167973

ABSTRACT

The central nervous system manifestations of systemic lupus erythematosus (SLE) remain poorly understood. Given the well-defined role of autoantibodies in other lupus manifestations, extensive work has gone into the identification of neuropathic autoantibodies. However, attempts to translate these findings to patients with SLE have yielded mixed results. We used the MRL/MpJ-Faslpr/lpr mouse, a well-established, spontaneous model of SLE, to establish the immune effectors responsible for brain disease. Transcriptomic analysis of the MRL/MpJ-Faslpr/lpr choroid plexus revealed an expression signature driving tertiary lymphoid structure formation, including chemokines related to stromal reorganization and lymphocyte compartmentalization. Additionally, transcriptional profiles indicated various stages of lymphocyte activation and germinal center formation. The extensive choroid plexus infiltrate present in MRL/MpJ-Faslpr/lpr mice with overt neurobehavioral deficits included locally proliferating B and T cells, intercellular interactions between lymphocytes and antigen-presenting cells, as well as evidence for in situ somatic hypermutation and class switch recombination. Furthermore, the choroid plexus was a site for trafficking lymphocytes into the brain. Finally, histological evaluation in human lupus patients with neuropsychiatric manifestations revealed increased leukocyte migration through the choroid plexus. These studies identify a potential new pathway underlying neuropsychiatric lupus and support tertiary lymphoid structure formation in the choroid plexus as a novel mechanism of brain-immune interfacing.


Subject(s)
Choroid Plexus , Lupus Vasculitis, Central Nervous System , Tertiary Lymphoid Structures , Animals , Choroid Plexus/metabolism , Choroid Plexus/pathology , Choroid Plexus/physiopathology , Disease Models, Animal , Female , Lupus Vasculitis, Central Nervous System/metabolism , Lupus Vasculitis, Central Nervous System/pathology , Lupus Vasculitis, Central Nervous System/physiopathology , Mice , Mice, Inbred MRL lpr , Tertiary Lymphoid Structures/metabolism , Tertiary Lymphoid Structures/pathology , Tertiary Lymphoid Structures/physiopathology , Transcriptome
11.
Am J Psychiatry ; 176(7): 564-572, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31164007

ABSTRACT

OBJECTIVE: The choroid plexus is an important physiological barrier and produces CSF and neurotrophic, angiogenic, and inflammatory factors involved in brain development. Choroid plexus abnormalities have been implicated in both schizophrenia and bipolar disorder. A previous choroid plexus transcriptomic analysis of schizophrenia identified an upregulation of immune and inflammatory genes that correlated with peripheral inflammatory markers. The purpose of this study was to examine choroid plexus volume in probands across the psychosis spectrum and in their first-degree and axis II cluster A relatives, as well as choroid plexus familiality and choroid plexus covariance with clinical, cognitive, brain, and peripheral marker measures. METHODS: Choroid plexus volume was quantified (using FreeSurfer) in psychosis probands, their first-degree and axis II cluster A relatives, and healthy control subjects, organized by DSM-IV-TR diagnosis. Analyte, structural connectivity, and genotype data were collected from a subset of study subjects. RESULTS: Choroid plexus volume was significantly larger in probands compared with first-degree relatives or healthy control subjects; first-degree relatives had intermediate enlargement compared with healthy control subjects; and total choroid plexus volume was significantly heritable. Larger volume was associated with worse cognition, smaller total gray matter and amygdala volume, larger lateral ventricle volume, and lower structural connectivity in probands. Associations between larger volume and higher levels of interleukin 6 in probands was also observed. CONCLUSIONS: These findings suggest the involvement of the choroid plexus across the psychosis spectrum with a potential pathophysiological mechanism involving the neuroimmune axis, which functions in maintaining brain homeostasis and interacting with the peripheral immune and inflammatory system. The choroid plexus may be an important target in future research.


Subject(s)
Choroid Plexus/pathology , Cognition , Inflammation/pathology , Psychotic Disorders/pathology , Adult , Case-Control Studies , Choroid Plexus/diagnostic imaging , Choroid Plexus/physiopathology , Cognition/physiology , Female , Humans , Inflammation/physiopathology , Magnetic Resonance Imaging , Male , Neuroimaging , Organ Size , Phenotype , Psychotic Disorders/diagnostic imaging , Psychotic Disorders/physiopathology
12.
Sci Adv ; 5(5): eaav4111, 2019 05.
Article in English | MEDLINE | ID: mdl-31149632

ABSTRACT

The immune system supports brain plasticity and homeostasis, yet it is prone to changes following psychological stress. Thus, it remains unclear whether and how stress-induced immune alterations contribute to the development of mental pathologies. Here, we show that following severe stress in mice, leukocyte trafficking through the choroid plexus (CP), a compartment that mediates physiological immune-brain communication, is impaired. Blocking glucocorticoid receptor signaling, either systemically or locally through its genetic knockdown at the CP, facilitated the recruitment of Gata3- and Foxp3-expressing T cells to the brain and attenuated post-traumatic behavioral deficits. These findings functionally link post-traumatic stress behavior with elevated stress-related corticosteroid signaling at the brain-immune interface and suggest a novel therapeutic target to attenuate the consequences of severe psychological stress.


Subject(s)
Adrenal Cortex Hormones/metabolism , Brain/immunology , Stress, Psychological/metabolism , Adrenal Cortex Hormones/cerebrospinal fluid , Adrenal Cortex Hormones/immunology , Animals , Behavior, Animal , Brain/metabolism , Choroid Plexus/metabolism , Choroid Plexus/physiopathology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , GATA3 Transcription Factor/metabolism , Hormone Antagonists/pharmacology , Humans , Mice, Inbred C57BL , Mice, Mutant Strains , Mifepristone/pharmacology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Signal Transduction , Single-Cell Analysis , Stress, Psychological/immunology , T-Lymphocytes/immunology
13.
Genet Med ; 21(3): 572-579, 2019 03.
Article in English | MEDLINE | ID: mdl-29907796

ABSTRACT

PURPOSE: SMARCB1 encodes a subunit of the SWI/SNF complex involved in chromatin remodeling. Pathogenic variants (PV) in this gene can give rise to three conditions. Heterozygous loss-of-function germline PV cause rhabdoid tumor predisposition syndrome and schwannomatosis. Missense PV and small in-frame deletions in exons 8 and 9 result in Coffin-Siris syndrome, which is characterized by intellectual disability (ID), coarse facial features, and fifth digit anomalies. METHODS: By a gene matching approach, individuals with a similar SMARCB1 PV were identified. Informed consent was obtained and patient data were collected to further establish genotype-phenotype relationship. RESULTS: A recurrent de novo missense PV (c.110G>A;p.Arg37His) in exon 2 of SMARCB1, encoding the DNA-binding domain, was identified in four individuals from different genetic centers. They shared a distinct phenotype consisting of profound ID and hydrocephalus due to choroid plexus hyperplasia. Other shared features include severe neonatal feeding difficulties; congenital heart, kidney, and eye anomalies; obstructive sleep apnea; and anemia. CONCLUSION: The p.Arg37His PV in the DNA-binding domain of SMARCB1 causes a distinctive syndrome, likely through a gain-of-function or dominant-negative effect, which is characterized by severe ID and hydrocephalus resulting from choroid plexus hyperplasia. This report broadens the phenotypic spectrum associated with PV in SMARCB1.


Subject(s)
Hydrocephalus/genetics , Intellectual Disability/genetics , SMARCB1 Protein/genetics , Adolescent , Child , Child, Preschool , Choroid Plexus/physiopathology , Chromatin Assembly and Disassembly/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Exome , Facies , Female , Genetic Association Studies , Humans , Hyperplasia/genetics , Infant , Male , Nuclear Proteins/genetics , Phenotype , SMARCB1 Protein/physiology , Transcription Factors/genetics
14.
EMBO Mol Med ; 11(1)2019 01.
Article in English | MEDLINE | ID: mdl-30518636

ABSTRACT

Though congenital hydrocephalus is heritable, it has been linked only to eight genes, one of which is MPDZ Humans and mice that carry a truncated version of MPDZ incur severe hydrocephalus resulting in acute morbidity and lethality. We show by magnetic resonance imaging that contrast medium penetrates into the brain ventricles of mice carrying a Mpdz loss-of-function mutation, whereas none is detected in the ventricles of normal mice, implying that the permeability of the choroid plexus epithelial cell monolayer is abnormally high. Comparative proteomic analysis of the cerebrospinal fluid of normal and hydrocephalic mice revealed up to a 53-fold increase in protein concentration, suggesting that transcytosis through the choroid plexus epithelial cells of Mpdz KO mice is substantially higher than in normal mice. These conclusions are supported by ultrastructural evidence, and by immunohistochemistry and cytology data. Our results provide a straightforward and concise explanation for the pathophysiology of Mpdz-linked hydrocephalus.


Subject(s)
Capillary Permeability , Carrier Proteins/genetics , Choroid Plexus/pathology , Choroid Plexus/physiopathology , Hydrocephalus/pathology , Hydrocephalus/physiopathology , Animals , Contrast Media/analysis , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Magnetic Resonance Imaging , Membrane Proteins , Mice
15.
J Am Heart Assoc ; 7(3)2018 01 31.
Article in English | MEDLINE | ID: mdl-29386206

ABSTRACT

BACKGROUND: Germinal matrix hemorrhage (GMH) is a leading cause of mortality and lifelong morbidity in preterm infants. Posthemorrhagic hydrocephalus (PHH) is a common complication of GMH. A sodium-coupled bicarbonate exchanger (NCBE) encoded by solute carrier family 4 member 10 gene is expressed on the choroid plexus basolateral membrane and may play a role in cerebrospinal fluid production and the development of PHH. Following GMH, iron degraded from hemoglobin has been linked to PHH. Choroid plexus epithelial cells also contain iron-responsive element-binding proteins (IRPs), IRP1, and IRP2 that bind to mRNA iron-responsive elements. The present study aims to resolve the following issues: (1) whether the expression of NCBE is regulated by IRPs; (2) whether NCBE regulates the formation of GMH-induced hydrocephalus; and (3) whether inhibition of NCBE reduces PHH development. METHODS AND RESULTS: GMH model was established in P7 rat pups by injecting bacterial collagenase into the right ganglionic eminence. Another group received iron trichloride injections instead of collagenase. Deferoxamine was administered intraperitoneally for 3 consecutive days after GMH/iron trichloride. Solute carrier family 4 member 10 small interfering RNA or scrambled small interfering RNA was administered by intracerebroventricular injection 24 hours before GMH and followed with an injection every 7 days over 21 days. NCBE expression increased while IRP2 expression decreased after GMH/iron trichloride. Deferoxamine ameliorated both the GMH-induced and iron trichloride-induced decrease of IRP2 and decreased NCBE expressions. Deferoxamine and solute carrier family 4 member 10 small interfering RNA improved cognitive and motor functions at 21 to 28 days post GMH and reduced cerebrospinal fluid production as well as the degree of hydrocephalus at 28 days after GMH. CONCLUSIONS: Targeting iron-induced overexpression of NCBE may be a translatable therapeutic strategy for the treatment of PHH following GMH.


Subject(s)
Cerebral Hemorrhage/therapy , Choroid Plexus/drug effects , Deferoxamine/pharmacology , Hydrocephalus/prevention & control , RNA, Small Interfering/administration & dosage , RNAi Therapeutics , Siderophores/pharmacology , Sodium-Bicarbonate Symporters/metabolism , Animals , Animals, Newborn , Behavior, Animal/drug effects , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/physiopathology , Cerebrospinal Fluid/metabolism , Chlorides , Choroid Plexus/metabolism , Choroid Plexus/physiopathology , Cognition/drug effects , Disease Models, Animal , Ferric Compounds , Hydrocephalus/genetics , Hydrocephalus/metabolism , Hydrocephalus/physiopathology , Injections, Intraventricular , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 1/metabolism , Iron Regulatory Protein 2/genetics , Iron Regulatory Protein 2/metabolism , Motor Activity/drug effects , Rats, Sprague-Dawley , Sodium-Bicarbonate Symporters/genetics
16.
Acta Neuropathol ; 134(6): 851-868, 2017 12.
Article in English | MEDLINE | ID: mdl-28762187

ABSTRACT

Neuroinflammation contributes substantially to stroke pathophysiology. Cerebral invasion of peripheral leukocytes-particularly T cells-has been shown to be a key event promoting inflammatory tissue damage after stroke. While previous research has focused on the vascular invasion of T cells into the ischemic brain, the choroid plexus (ChP) as an alternative cerebral T-cell invasion route after stroke has not been investigated. We here report specific accumulation of T cells in the peri-infarct cortex and detection of T cells as the predominant population in the ipsilateral ChP in mice as well as in human post-stroke autopsy samples. T-cell migration from the ChP to the peri-infarct cortex was confirmed by in vivo cell tracking of photoactivated T cells. In turn, significantly less T cells invaded the ischemic brain after photothrombotic lesion of the ipsilateral ChP and in a stroke model encompassing ChP ischemia. We detected a gradient of CCR2 ligands as the potential driving force and characterized the neuroanatomical pathway for the intracerebral migration. In summary, our study demonstrates that the ChP is a key invasion route for post-stroke cerebral T-cell invasion and describes a CCR2-ligand gradient between cortex and ChP as the potential driving mechanism for this invasion route.


Subject(s)
Brain Ischemia/physiopathology , Cell Movement/physiology , Choroid Plexus/physiopathology , Stroke/physiopathology , T-Lymphocytes/physiology , Aged , Aged, 80 and over , Animals , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Brain Ischemia/pathology , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Chemokine CCL2/metabolism , Choroid Plexus/pathology , Disease Models, Animal , Female , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/pathology , Myeloid Cells/physiology , Stroke/pathology , T-Lymphocytes/pathology
17.
Fluids Barriers CNS ; 14(1): 8, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28351417

ABSTRACT

While the impact of hemorrhagic and ischemic strokes on the blood-brain barrier has been extensively studied, the impact of these types of stroke on the choroid plexus, site of the blood-CSF barrier, has received much less attention. The purpose of this review is to examine evidence of choroid plexus injury in clinical and preclinical studies of intraventricular hemorrhage, subarachnoid hemorrhage, intracerebral hemorrhage and ischemic stroke. It then discusses evidence that the choroid plexuses are important in the response to brain injury, with potential roles in limiting damage. The overall aim of the review is to highlight deficiencies in our knowledge on the impact of hemorrhagic and ischemic strokes on the choroid plexus, particularly with reference to intraventricular hemorrhage, and to suggest that a greater understanding of the response of the choroid plexus to stroke may open new avenues for brain protection.


Subject(s)
Brain Ischemia/physiopathology , Cerebral Hemorrhage/physiopathology , Choroid Plexus/physiopathology , Stroke/physiopathology , Animals , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Brain Ischemia/pathology , Brain Ischemia/therapy , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/therapy , Choroid Plexus/pathology , Humans , Neuroprotection/physiology , Stroke/pathology , Stroke/therapy
18.
Neurobiol Dis ; 107: 32-40, 2017 Nov.
Article in English | MEDLINE | ID: mdl-27546055

ABSTRACT

This article brings the choroid plexus into the context of health and disease. It is remarkable that the choroid plexus, composed by a monolayer of epithelial cells that lie in a highly vascularized stroma, floating within the brain ventricles, gets so little attention in major physiology and medicine text books and in the scientific literature in general. Consider that it is responsible for producing most of the about 150mL of cerebrospinal fluid that fills the brain ventricles and the subarachnoid space and surrounds the spinal cord in the adult human central nervous system, which is renewed approximately 2-3 times daily. As such, its activity influences brain metabolism and function, which will be addressed. Reflect that it contains an impressive number of receptors and transporters, both in the apical and basolateral sides of the epithelial cells, and as such is a key structure for the communication between the brain and the periphery. This will be highlighted in the context of neonatal jaundice, multiple sclerosis and Alzheimer's disease. Realize that the capillaries that irrigate the choroid plexus stroma do not possess tight junctions and that the blood flow to the choroid plexus is five times higher than that in the brain parenchyma, allowing for a rapid sensing system and delivery of molecules such as nutrients and metals as will be revised. Recognize that certain drugs reach the brain parenchyma solely through the choroid plexus epithelia, which has potential to be manipulated in diseases such as neonatal jaundice and Alzheimer's disease as will be discussed. Without further notice, it must be now clear that understanding the choroid plexus is necessary for comprehending the brain and how the brain is modulated and modulates all other systems, in health and in disease. This review article intends to address current knowledge on the choroid plexus, and to motivate the scientific community to consider it when studying normal brain physiology and diseases of the central nervous system. It will guide the reader through several aspects of the choroid plexus in normal physiology, in diseases characteristic of various periods of life (newborns-kernicterus, young adults-multiple sclerosis and the elder-Alzheimer's disease), and how sex-differences may relate to disease susceptibility.


Subject(s)
Choroid Plexus/physiology , Choroid Plexus/physiopathology , Animals , Choroid Plexus/anatomy & histology , Humans
19.
Sci Rep ; 6: 39070, 2016 12 12.
Article in English | MEDLINE | ID: mdl-27941913

ABSTRACT

To clarify the pathogenesis of two different types of adult-onset normal-pressure hydrocephalus (NPH), we investigated cerebrospinal fluid distribution on the high-field three-dimensional MRI. The subarachnoid spaces in secondary NPH were smaller than those in the controls, whereas those in idiopathic NPH were of similar size to the controls. In idiopathic NPH, however, the basal cistern and Sylvian fissure were enlarged in concurrence with ventricular enlargement towards the z-direction, but the convexity subarachnoid space was severely diminished. In this article, we provide evidence that the key cause of the disproportionate cerebrospinal fluid distribution in idiopathic NPH is the compensatory direct CSF communication between the inferior horn of the lateral ventricles and the ambient cistern at the choroidal fissure. In contrast, all parts of the subarachnoid spaces were equally and severely decreased in secondary NPH. Blockage of CSF drainage from the subarachnoid spaces could cause the omnidirectional ventricular enlargement in secondary NPH.


Subject(s)
Cerebrospinal Fluid Leak/physiopathology , Choroid Plexus/abnormalities , Hydrocephalus, Normal Pressure/pathology , Lateral Ventricles/abnormalities , Subarachnoid Space/abnormalities , Age of Onset , Aged , Aged, 80 and over , Cerebrospinal Fluid Leak/diagnostic imaging , Choroid Plexus/diagnostic imaging , Choroid Plexus/physiopathology , Female , Humans , Hydrocephalus, Normal Pressure/classification , Hydrocephalus, Normal Pressure/diagnostic imaging , Hydrocephalus, Normal Pressure/physiopathology , Lateral Ventricles/diagnostic imaging , Lateral Ventricles/physiopathology , Magnetic Resonance Imaging/methods , Male , Subarachnoid Space/diagnostic imaging , Subarachnoid Space/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...