Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Fluids Barriers CNS ; 19(1): 47, 2022 Jun 07.
Article in English | MEDLINE | ID: mdl-35672829

ABSTRACT

BACKGROUND: Severe neonatal jaundice resulting from elevated levels of unconjugated bilirubin in the blood induces dramatic neurological impairment. Central oxidative stress and an inflammatory response have been associated with the pathophysiological mechanism. Cells forming the blood-brain barrier and the choroidal blood-CSF barrier are the first CNS cells exposed to increased plasma levels of unconjugated bilirubin. These barriers are key regulators of brain homeostasis and require active oxidative metabolism to fulfill their protective functions. The choroid plexus-CSF system is involved in neuroinflammatory processes. In this paper, we address the impact of neonatal hyperbilirubinemia on some aspects of brain barriers. We describe physiological changes in the neurovascular network, blood-brain/CSF barriers integrities, and CSF cytokine levels during the postnatal period in normobilirubinemic animals, and analyze these parameters in parallel in Gunn rats that are deficient in bilirubin catabolism and develop postnatal hyperbilirubinemia. METHODS: Gunn rats bearing a mutation in UGT1a genes were used. The neurovascular network was analyzed by immunofluorescence stereomicroscopy. The integrity of the barriers was evaluated by [14C]-sucrose permeability measurement. CSF cytokine levels were measured by multiplex immunoassay. The choroid plexus-CSF system response to an inflammatory challenge was assessed by enumerating CSF leukocytes. RESULTS: In normobilirubinemic animals, the neurovascular network expands postnatally and displays stage-specific regional variations in its complexity. Network expansion is not affected by hyperbilirubinemia. Permeability of the blood-brain and blood-CSF barriers to sucrose decreases between one- and 9-day-old animals, and does not differ between normobilirubinemic and hyperbilirubinemic rats. Cytokine profiles differ between CSF and plasma in all 1-, 9-, and 18-day-old animals. The CSF cytokine profile in 1-day-old animals is markedly different from that established in older animals. Hyperbilirubinemia perturbs these cytokine profiles only to a very limited extent, and reduces CSF immune cell infiltration triggered by systemic exposure to a bacterial lipopeptide. CONCLUSION: The data highlight developmental specificities of the blood-brain barrier organization and of CSF cytokine content. They also indicate that a direct effect of bilirubin on the vascular system organization, brain barriers morphological integrity, and inflammatory response of the choroid plexus-CSF system is not involved in the alteration of brain functions induced by severe neonatal jaundice.


Subject(s)
Blood-Brain Barrier , Jaundice, Neonatal , Animals , Bilirubin/metabolism , Blood-Brain Barrier/metabolism , Cerebrospinal Fluid/metabolism , Choroid Plexus/metabolism , Cytokines/metabolism , Humans , Hyperbilirubinemia/metabolism , Infant, Newborn , Jaundice, Neonatal/metabolism , Rats , Rats, Gunn , Sucrose
2.
PLoS One ; 15(9): e0238301, 2020.
Article in English | MEDLINE | ID: mdl-32881954

ABSTRACT

BACKGROUND: Neuromyelitis optica spectrum disorders (NMOSD) is a primary astrocytopathy driven by antibodies directed against the aquaporin-4 water channel located at the end-feet of the astrocyte. Although blood-brain barrier (BBB) breakdown is considered one of the key steps for the development and lesion formation, little is known about the molecular mechanisms involved. The aim of the study was to evaluate the effect of human immunoglobulins from NMOSD patients (NMO-IgG) on BBB properties. METHODS: Freshly isolated brain microvessels (IBMs) from rat brains were used as a study model. At first, analysis of the secretome profile from IBMs exposed to purified NMO-IgG, to healthy donor IgG (Control-IgG), or non-treated, was performed. Second, tight junction (TJ) proteins expression in fresh IBMs and primary cultures of brain microvascular endothelial cells (BMEC) was analysed by Western blotting (Wb) after exposition to NMO-IgG and Control-IgG. Finally, functional BBB properties were investigated evaluating the presence of rat-IgG in tissue lysate from brain using Wb in the rat-model, and the passage of NMO-IgG and sucrose in a bicameral model. RESULTS: We found that NMO-IgG induces functional and morphological BBB changes, including: 1) increase of pro-inflammatory cytokines production (CXCL-10 [IP-10], IL-6, IL-1RA, IL-1ß and CXCL-3) in IBMs when exposed to NMO-IgG; 2) decrease of Claudin-5 levels by 25.6% after treatment of fresh IBMs by NMO-IgG compared to Control-IgG (p = 0.002), and similarly, decrease of Claudin-5 by at least 20% when BMEC were cultured with NMO-IgG from five different patients; 3) a higher level of rat-IgG accumulated in periventricular regions of NMO-rats compared to Control-rats and an increase in the permeability of BBB after NMO-IgG treatment in the bicameral model. CONCLUSION: Human NMO-IgG induces both structural and functional alterations of BBB properties, suggesting a direct role of NMO-IgG on modulation of BBB permeability in NMOSD.


Subject(s)
Aquaporin 4/immunology , Blood-Brain Barrier/metabolism , Immunoglobulin G/pharmacology , Neuromyelitis Optica/pathology , Permeability/drug effects , Animals , Blood-Brain Barrier/drug effects , Cells, Cultured , Chemokines/metabolism , Claudin-5/metabolism , Cytokines/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Immunoglobulin G/isolation & purification , Microvessels/cytology , Microvessels/metabolism , Neuromyelitis Optica/metabolism , Rats
3.
Acta Neuropathol Commun ; 8(1): 4, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31973769

ABSTRACT

The etiology of neurological impairments associated with prematurity and other perinatal complications often involves an infectious or pro-inflammatory component. The use of antioxidant molecules have proved useful to protect the neonatal brain from injury. The choroid plexuses-CSF system shapes the central nervous system response to inflammation at the adult stage, but little is known on the neuroimmune interactions that take place at the choroidal blood-CSF barrier during development. We previously described that peripheral administration to neonatal mice of the TLR2 ligand PAM3CSK4 (P3C), a prototypic Gram-positive bacterial lipopeptide, induces the migration of innate immune cells to the CSF. Here we showed in neonatal rats exposed to P3C that the migration of neutrophils into the CSF, which occurred through the choroid plexuses, is abolished following administration of the antioxidant drug N-acetylcysteine. Combining light sheet microscopy imaging of choroid plexus, a differentiated model of the blood-CSF barrier, and multiplex cytokine assays, we showed that the choroidal epithelium responds to the bacterial insult by a specific pattern of cytokine secretion, leading to a selective accumulation of neutrophils in the choroid plexus and to their trafficking into CSF. N-acetylcysteine acted by blocking neutrophil migration across both the endothelium of choroidal stromal vessels and the epithelium forming the blood-CSF barrier, without interfering with neutrophil blood count, neutrophil tropism for choroid plexus, and choroidal chemokine-driven chemotaxis. N-acetylcysteine reduced the injury induced by hypoxia-ischemia in P3C-sensitized neonatal rats. Overall, the data show that a double endothelial and epithelial check point controls the transchoroidal migration of neutrophils into the developing brain. They also point to the efficacy of N-acetylcysteine in reducing the deleterious effects of inflammation-associated perinatal injuries by a previously undescribed mechanism, i.e. the inhibition of innate immune cell migration across the choroid plexuses, without interfering with the systemic inflammatory response to infection.


Subject(s)
Acetylcysteine/administration & dosage , Antioxidants/administration & dosage , Brain/immunology , Cell Movement/drug effects , Cerebrospinal Fluid/immunology , Choroid Plexus/immunology , Lipopeptides/administration & dosage , Neutrophils/immunology , Animals , Brain/drug effects , Brain/growth & development , Cells, Cultured , Choroid Plexus/drug effects , Female , Inflammation Mediators/immunology , Leukocytes/immunology , Neutrophils/drug effects , Rats, Sprague-Dawley , Rats, Wistar
4.
J Neurosci ; 38(14): 3466-3479, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29507144

ABSTRACT

Exposure of the developing brain to toxins, drugs, or deleterious endogenous compounds during the perinatal period can trigger alterations in cell division, migration, differentiation, and synaptogenesis, leading to lifelong neurological impairment. The brain is protected by cellular barriers acting through multiple mechanisms, some of which are still poorly explored. We used a combination of enzymatic assays, live tissue fluorescence microscopy, and an in vitro cellular model of the blood-CSF barrier to investigate an enzymatic detoxification pathway in the developing male and female rat brain. We show that during the early postnatal period the choroid plexus epithelium forming the blood-CSF barrier and the ependymal cell layer bordering the ventricles harbor a high detoxifying capacity that involves glutathione S-transferases. Using a functional knock-down rat model for choroidal glutathione conjugation, we demonstrate that already in neonates, this metabolic pathway efficiently prevents the penetration of blood-borne reactive compounds into CSF. The versatility of the protective mechanism results from the multiplicity of the glutathione S-transferase isoenzymes, which are differently expressed between the choroidal epithelium and the ependyma. The various isoenzymes display differential substrate specificities, which greatly widen the spectrum of molecules that can be inactivated by this pathway. In conclusion, the blood-CSF barrier and the ependyma are identified as key cellular structures in the CNS to protect the brain fluid environment from different chemical classes of potentially toxic compounds during the postnatal period. This metabolic neuroprotective function of brain interfaces ought to compensate for the liver postnatal immaturity.SIGNIFICANCE STATEMENT Brain homeostasis requires a stable and controlled internal environment. Defective brain protection during the perinatal period can lead to lifelong neurological impairment. We demonstrate that the choroid plexus forming the blood-CSF barrier is a key player in the protection of the developing brain. Glutathione-dependent enzymatic metabolism in the choroidal epithelium inactivates a broad spectrum of noxious compounds, efficiently preventing their penetration into the CSF. A second line of detoxification is located in the ependyma separating the CSF from brain tissue. Our study reveals a novel facet of the mechanisms by which the brain is protected at a period of high vulnerability, at a time when the astrocytic network is still immature and liver xenobiotic metabolism is limited.


Subject(s)
Blood-Brain Barrier/metabolism , Glutathione Transferase/metabolism , Glutathione/metabolism , Animals , Blood-Brain Barrier/growth & development , Choroid Plexus/growth & development , Choroid Plexus/metabolism , Ependyma/growth & development , Ependyma/metabolism , Female , Free Radicals/blood , Free Radicals/cerebrospinal fluid , Glutathione/blood , Glutathione/cerebrospinal fluid , Male , Rats , Rats, Sprague-Dawley
5.
Eur J Med Chem ; 98: 237-49, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26043161

ABSTRACT

Cycloaddition between (+) or (-)-menthone-derived nitrones and N-benzyl-3-pyrroline afforded enantiopure spiro-fused heterocycles. The reaction occurred enantio- and diastereo-selectively on the less hindered side of the nitrone, the 3-pyrroline N-benzyl group being oriented outwards, thus controlling the configurations of three simultaneously created chiral centers. From either (+) or (-)-menthone, both enantiomeric cycloadducts were synthesized in excellent yield. Removing the chiral auxiliary and the N-benzyl group delivered a series of enantiopure 4-hydroxy-3-glycinyl-pyrrolidine derivatives in 3-5 steps and 36 to 81 overall yields. Using two other achiral nitrones, shorter routes to racemic analogues were developed. Two of the synthesized compounds markedly lowered extracellular glutamate level and modestly interacted with cannabinoid type-1 receptors. As these two neuroactive compounds were devoid of in vitro toxicity and did not cross the blood brain interface, they might represent potential pharmacological agents to target peripheral organs.


Subject(s)
Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Acetates/chemistry , Animals , Drug Evaluation, Preclinical , Male , Models, Molecular , Rats , Rats, Wistar , Stereoisomerism
6.
Fluids Barriers CNS ; 12: 8, 2015.
Article in English | MEDLINE | ID: mdl-25793105

ABSTRACT

BACKGROUND: The cerebrospinal fluid (CSF) circulatory system is involved in neuroimmune regulation, cerebral detoxification, and delivery of various endogenous and exogenous substances. In conjunction with the choroid plexuses, which form the main barrier site between blood and CSF, this fluid participates in controlling the environment of the developing brain. The lack of comprehensive data on developmental changes in CSF volume and distribution impairs our understanding of CSF contribution to brain development, and limits the interpretation of blood-CSF permeability data. To address these issues, we describe the evolution of the CSF circulatory system during the perinatal period and have quantified the volume of the different ventricular, cisternal and subarachnoid CSF compartments at three ages in developing rats. METHODS: Immunohistofluorescence was used to visualize tight junctions in parenchymal and meningeal vessels, and in choroid plexus epithelium of 19-day fetal rats. A quantitative method based on serial sectioning of frozen head and surface measurements at the cutting plane was used to determine the volume of twenty different CSF compartments in rat brain on embryonic day 19 (E19), and postnatal days 2 (P2) and 9 (P9). Blood-CSF permeability constants for sucrose were established at P2 and P9, following CSF sampling from the cisterna magna. RESULTS: Claudin-1 and claudin-5 immunohistofluorescence labeling illustrated the barrier phenotype acquired by all blood-brain and blood-CSF interfaces throughout the entire CNS in E19 rats. This should ensure that brain fluid composition is regulated and independent from plasma composition in developing brain. Analysis of the caudo-rostral profiles of CSF distribution and of the volume of twenty CSF compartments indicated that the CSF-to-cranial cavity volume ratio decreases from 30% at E19 to 10% at P9. CSF compartmentalization within the brain changes during this period, with a major decrease in CSF-to-brain volume ratio in the caudal half of the brain. Integrating CSF volume with the measurement of permeability constants, adds to our understanding of the apparent postnatal decrease in blood-CSF permeability to sucrose. CONCLUSION: Reference data on CSF compartment volumes throughout development are provided. Such data can be used to refine blood-CSF permeability constants in developing rats, and should help a better understanding of diffusion, bulk flow, and volume transmission in the developing brain.

7.
Mol Pharm ; 8(3): 651-63, 2011 Jun 06.
Article in English | MEDLINE | ID: mdl-21438632

ABSTRACT

Brain penetration is characterized by its extent and rate and is influenced by drug physicochemical properties, plasma exposure, plasma and brain protein binding and BBB permeability. This raises questions related to physiology, interspecies differences and in vitro/in vivo extrapolation. We herein discuss the use of in vitro human and animal BBB model as a tool to improve CNS compound selection. These cell-based BBB models are characterized by low paracellular permeation, well-developed tight junctions and functional efflux transporters. A study of twenty drugs shows similar compound ranking between rat and human models although with a 2-fold higher permeability in rat. cLogP < 5, PSA < 120 Å, MW < 450 were confirmed as essential for CNS drugs. An in vitro/in vivo correlation in rat (R² = 0.67; P = 2 × 10⁻4) was highlighted when in vitro permeability and efflux were considered together with plasma exposure and free fraction. The cell-based BBB model is suitable to optimize CNS-drug selection, to study interspecies differences and then to support human brain exposure prediction.


Subject(s)
Blood-Brain Barrier/metabolism , Animals , Biological Transport/physiology , Brain/metabolism , Cells, Cultured , Drug Discovery/methods , Humans , Male , Models, Biological , Rats , Tight Junctions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...