Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 38(14): 3466-3479, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29507144

RESUMEN

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.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Glutatión Transferasa/metabolismo , Glutatión/metabolismo , Animales , Barrera Hematoencefálica/crecimiento & desarrollo , Plexo Coroideo/crecimiento & desarrollo , Plexo Coroideo/metabolismo , Epéndimo/crecimiento & desarrollo , Epéndimo/metabolismo , Femenino , Radicales Libres/sangre , Radicales Libres/líquido cefalorraquídeo , Glutatión/sangre , Glutatión/líquido cefalorraquídeo , Masculino , Ratas , Ratas Sprague-Dawley
2.
Am J Physiol Cell Physiol ; 315(4): C445-C456, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29949405

RESUMEN

Hydrogen peroxide, released at low physiological concentration, is involved in different cell signaling pathways during brain development. When released at supraphysiological concentrations in brain fluids following an inflammatory, hypoxic, or toxic stress, it can initiate lipid peroxidation, protein, and nucleic acid damage and contribute to long-term neurological impairment associated with perinatal diseases. We found high glutathione peroxidase and glutathione reductase enzymatic activities in both lateral and fourth ventricle choroid plexus tissue isolated from developing rats, in comparison to the cerebral cortex and liver. Consistent with these, a high protein expression of glutathione peroxidases 1 and 4 was observed in choroid plexus epithelial cells, which form the blood-cerebrospinal fluid barrier. Live choroid plexuses isolated from newborn rats were highly efficient in detoxifying H2O2 from mock cerebrospinal fluid, illustrating the capacity of the choroid plexuses to control H2O2 concentration in the ventricular system of the brain. We used a differentiated cellular model of the blood-cerebrospinal fluid barrier coupled to kinetic and inhibition analyses to show that glutathione peroxidases are more potent than catalase to detoxify extracellular H2O2 at concentrations up to 250 µM. The choroidal cells also formed an enzymatic barrier preventing blood-borne hydroperoxides to reach the cerebrospinal fluid. These data point out the choroid plexuses as key structures in the control of hydroperoxide levels in the cerebral fluid environment during development, at a time when the protective glial cell network is still immature. Glutathione peroxidases are the main effectors of this choroidal hydroperoxide inactivation.


Asunto(s)
Encéfalo/metabolismo , Líquido Cefalorraquídeo/metabolismo , Plexo Coroideo/metabolismo , Peróxido de Hidrógeno/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Células Epiteliales/metabolismo , Femenino , Masculino , Ratas , Ratas Sprague-Dawley
3.
Acta Neuropathol ; 135(3): 337-361, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29368213

RESUMEN

The barrier between the blood and the ventricular cerebrospinal fluid (CSF) is located at the choroid plexuses. At the interface between two circulating fluids, these richly vascularized veil-like structures display a peculiar morphology explained by their developmental origin, and fulfill several functions essential for CNS homeostasis. They form a neuroprotective barrier preventing the accumulation of noxious compounds into the CSF and brain, and secrete CSF, which participates in the maintenance of a stable CNS internal environment. The CSF circulation plays an important role in volume transmission within the developing and adult brain, and CSF compartments are key to the immune surveillance of the CNS. In these contexts, the choroid plexuses are an important source of biologically active molecules involved in brain development, stem cell proliferation and differentiation, and brain repair. By sensing both physiological changes in brain homeostasis and peripheral or central insults such as inflammation, they also act as sentinels for the CNS. Finally, their role in the control of immune cell traffic between the blood and the CSF confers on the choroid plexuses a function in neuroimmune regulation and implicates them in neuroinflammation. The choroid plexuses, therefore, deserve more attention while investigating the pathophysiology of CNS diseases and related comorbidities.


Asunto(s)
Barrera Hematoencefálica/anatomía & histología , Barrera Hematoencefálica/metabolismo , Ventrículos Cerebrales/anatomía & histología , Ventrículos Cerebrales/metabolismo , Líquido Cefalorraquídeo/metabolismo , Animales , Barrera Hematoencefálica/patología , Ventrículos Cerebrales/patología , Humanos , Neuroprotección/fisiología
4.
Pharm Res ; 35(4): 84, 2018 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-29516182

RESUMEN

Drug bioavailability to the developing brain is a major concern in the treatment of neonates and infants as well as pregnant and breast-feeding women. Central adverse drug reactions can have dramatic consequences for brain development, leading to major neurological impairment. Factors setting the cerebral bioavailability of drugs include protein-unbound drug concentration in plasma, local cerebral blood flow, permeability across blood-brain interfaces, binding to neural cells, volume of cerebral fluid compartments, and cerebrospinal fluid secretion rate. Most of these factors change during development, which will affect cerebral drug concentrations. Regarding the impact of blood-brain interfaces, the blood-brain barrier located at the cerebral endothelium and the blood-cerebrospinal fluid barrier located at the choroid plexus epithelium both display a tight phenotype early on in embryos. However, the developmental regulation of some multispecific efflux transporters that also limit the entry of numerous drugs into the brain through barrier cells is expected to favor drug penetration in the neonatal brain. Finally, drug cerebral bioavailability is likely to be affected following perinatal injuries that alter blood-brain interface properties. A thorough investigation of these mechanisms is mandatory for a better risk assessment of drug treatments in pregnant or breast-feeding women, and in neonate and pediatric patients.


Asunto(s)
Anomalías Inducidas por Medicamentos/prevención & control , Encéfalo/efectos de los fármacos , Lactancia Materna , Intercambio Materno-Fetal/efectos de los fármacos , Complicaciones del Embarazo/tratamiento farmacológico , Anomalías Inducidas por Medicamentos/etiología , Disponibilidad Biológica , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Desarrollo Infantil/efectos de los fármacos , Femenino , Desarrollo Fetal/efectos de los fármacos , Humanos , Lactante , Recién Nacido , Embarazo , Medición de Riesgo , Distribución Tisular
5.
Fluids Barriers CNS ; 21(1): 66, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39152442

RESUMEN

BACKGROUND: Group B Streptococcus (GBS) is the leading cause of neonatal meningitis responsible for a substantial cause of death and disability worldwide. The vast majority of GBS neonatal meningitis cases are due to the CC17 hypervirulent clone. However, the cellular and molecular pathways involved in brain invasion by GBS CC17 isolates remain largely elusive. Here, we studied the specific interaction of the CC17 clone with the choroid plexus, the main component of the blood-cerebrospinal fluid (CSF) barrier. METHODS: The interaction of GBS CC17 or non-CC17 strains with choroid plexus cells was studied using an in vivo mouse model of meningitis and in vitro models of primary and transformed rodent choroid plexus epithelial cells (CPEC and Z310). In vivo interaction of GBS with the choroid plexus was assessed by microscopy. Bacterial invasion and cell barrier penetration were examined in vitro, as well as chemokines and cytokines in response to infection. RESULTS: GBS CC17 was found associated with the choroid plexus of the lateral, 3rd and 4th ventricles. Infection of choroid plexus epithelial cells revealed an efficient internalization of the bacteria into the cells with GBS CC17 displaying a greater ability to invade these cells than a non-CC17 strain. Internalization of the GBS CC17 strain involved the CC17-specific HvgA adhesin and occurred via a clathrin-dependent mechanism leading to transcellular transcytosis across the choroid plexus epithelial monolayer. CPEC infection resulted in the secretion of several chemokines, including CCL2, CCL3, CCL20, CX3CL1, and the matrix metalloproteinase MMP3, as well as immune cell infiltration. CONCLUSION: Our findings reveal a GBS strain-specific ability to infect the blood-CSF barrier, which appears to be an important site of bacterial entry and an active site of immune cell trafficking in response to infection.


Asunto(s)
Plexo Coroideo , Streptococcus agalactiae , Plexo Coroideo/metabolismo , Plexo Coroideo/microbiología , Plexo Coroideo/inmunología , Animales , Streptococcus agalactiae/patogenicidad , Ratones , Adhesinas Bacterianas/metabolismo , Virulencia , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Barrera Hematoencefálica/microbiología , Barrera Hematoencefálica/metabolismo , Modelos Animales de Enfermedad , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/inmunología , Ratones Endogámicos C57BL , Transcitosis/fisiología , Femenino
6.
J Neuroinflammation ; 9: 187, 2012 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-22870891

RESUMEN

BACKGROUND: Cerebrospinal fluid (CSF) has been considered as a preferential pathway of circulation for immune cells during neuroimmune surveillance. In order to evaluate the involvement of CSF-filled spaces in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis, we performed a time-course analysis of immune cell association with the CSF-containing ventricles, velae, and cisterns in two active models of this disease. METHODS: Guinea-pig spinal cord homogenate-induced EAE in rat and myelin oligodendrocyte glycoprotein-induced EAE in mouse were used. Leukocyte distribution and phenotypes were investigated by immunohistochemistry in serial sections of brain areas of interest, as well as in CSF withdrawn from rat. Immune cells associated with the choroid plexuses were quantified. RESULTS: Freund's adjuvant-induced peripheral inflammation in the absence of brain antigen led to a subtle but definite increase in the number of myeloid cells in the extraventricular CSF spaces. In both rats and mice, EAE was characterized by a sustained and initial infiltration of lymphocytes and monocytes within forebrain/midbrain fluid-filled compartments such as the velum interpositum and ambient cisterns, and certain basal cisterns. Leukocytes further infiltrated periventricular and pericisternal parenchymal areas, along perivascular spaces or following a downward CSF-to-tissue gradient. Cells quantified in CSF sampled from rats included lymphocytes and neutrophils. The distinctive pattern of cell distribution suggests that both the choroid plexus and the vessels lying in the velae and cisterns are gates for early leukocyte entry in the central nervous system. B-cell infiltration observed in the mouse model was restricted to CSF-filled extraventricular compartments. CONCLUSION: These results identified distinctive velae and cisterns of the forebrain and midbrain as preferential sites of immune cell homing following peripheral and early central inflammation and point to a role of CSF in directing brain invasion by immune cells during EAE.


Asunto(s)
Encefalomielitis Autoinmune Experimental/líquido cefalorraquídeo , Encefalomielitis Autoinmune Experimental/patología , Leucocitos/patología , Prosencéfalo/patología , Secuencia de Aminoácidos , Animales , Biomarcadores/líquido cefalorraquídeo , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Cobayas , Inflamación/líquido cefalorraquídeo , Inflamación/inmunología , Inflamación/patología , Leucocitos/inmunología , Leucocitos/metabolismo , Mesencéfalo/inmunología , Mesencéfalo/metabolismo , Mesencéfalo/patología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Prosencéfalo/metabolismo , Ratas , Transducción de Señal/inmunología
7.
Histochem Cell Biol ; 138(6): 861-79, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22886143

RESUMEN

The choroid plexus epithelium controls the movement of solutes between the blood and the cerebrospinal fluid. It has been considered as a functionally more immature interface during brain development than in adult. The anatomical basis of this barrier is the interepithelial choroidal junction whose tightness has been attributed to the presence of claudins. We used quantitative real-time polymerase chain reaction, Western blot and immunohistochemistry to identify different claudins in the choroid plexuses of developing and adult rats. Claudin-1, -2, and -3 were highly and selectively expressed in the choroid plexus as compared to brain or parenchyma microvessels and were localized at epithelial junctions. Claudin-6, -9, -19, and -22 also displayed a previously undescribed choroidal selectivity, while claudin-4, -5, and -16 were enriched in the cerebral microvessels. The choroidal pattern of tight junction protein expression in prenatal brains was already complex and included occludin and zonula occludens proteins. It differed from the adult pattern in that the pore-forming claudin-2, claudin-9, and claudin-22 increased during development, while claudin-3 and claudin-6 decreased. Claudin-2 and claudin-11 presented a mirror image of abundance between lateral ventricle and fourth ventricle choroid plexuses. Imunohistochemical analysis of human fetal and postnatal brains for claudin-1, -2, and -3 demonstrated their early presence and localization at the apico-lateral border of the choroid plexus epithelial cells. Overall, choroidal epithelial tight junctions are already complex in developing brain. The observed differences in claudin expression between developing and adult choroid plexuses may indicate developmental differences in selective blood-cerebrospinal fluid transport functions.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Claudinas/análisis , Claudinas/genética , Perfilación de la Expresión Génica , Animales , Western Blotting , Plexo Coroideo/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Uniones Estrechas/metabolismo
8.
Fluids Barriers CNS ; 19(1): 47, 2022 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672829

RESUMEN

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.


Asunto(s)
Barrera Hematoencefálica , Ictericia Neonatal , Animales , Bilirrubina/metabolismo , Barrera Hematoencefálica/metabolismo , Líquido Cefalorraquídeo/metabolismo , Plexo Coroideo/metabolismo , Citocinas/metabolismo , Humanos , Hiperbilirrubinemia/metabolismo , Recién Nacido , Ictericia Neonatal/metabolismo , Ratas , Ratas Gunn , Sacarosa
9.
J Neurochem ; 117(4): 747-56, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21395586

RESUMEN

Manganese is an essential trace element, and a contrast agent of potential interest for brain magnetic resonance imaging. Brain overexposure to manganese, however induces a neurodegenerative syndrome. Imaging data suggest that manganese appearance into the CSF precedes its accumulation into the cerebral parenchyma. We therefore investigated manganese uptake and transport at the blood-CSF barrier. Like lead, the non protein-bound divalent manganese accumulated into the rat choroid plexus. The metal accumulation was especially high in developing animals. Using a differentiated cellular model of the blood-CSF barrier, we demonstrated that manganese crosses the choroid plexus epithelium by a concentrating, unidirectional blood-to-CSF transport mechanism. This transport was inhibited by calcium, which is also transported into the CSF against its concentration gradient. The permeability barrier function towards lipid-insoluble compound and the organic anion transport property of the blood-brain interface were affected by exposure of the blood-facing membrane of choroidal cells to micromolar concentrations of manganese, but its antioxidant capacity was not. The unidirectional transport of manganese across the choroid plexus provides the anatomo-functional basis linking the systemic exposure to manganese with the spreading pattern of manganese accumulation observed in brain imaging, and explains the polarized sensitivity of choroidal epithelial cells to manganese toxicity.


Asunto(s)
Encéfalo/metabolismo , Manganeso/líquido cefalorraquídeo , Manganeso/metabolismo , Animales , Transporte Biológico Activo , Barrera Hematoencefálica , Calcio/metabolismo , Calcio/farmacología , Permeabilidad de la Membrana Celular , Células Cultivadas , Plexo Coroideo/metabolismo , Cisteína/metabolismo , Dinoprostona/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Glutatión/metabolismo , Indicadores y Reactivos , Masculino , Manganeso/sangre , Metales/metabolismo , Ratas , Ratas Sprague-Dawley , Sacarosa/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo
10.
J Neurotrauma ; 38(4): 385-398, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-32940130

RESUMEN

Neuroinflammation and dysfunction of the blood-brain barrier (BBB) are two prominent mechanisms of secondary injury in neurotrauma. It has been suggested that Toll-like receptors (TLRs) play important roles in initiating and propagating neuroinflammation resulting from traumatic brain injury (TBI), but potential beneficial effects of targeting these receptors in TBI have not been broadly studied. Here, we investigated the effect of targeting TLRs with proteoglycan 4 (PRG4) on post-traumatic neuroinflammation and BBB function. PRG4 is a mucinous glycoprotein with strong anti-inflammatory properties, exerting its biological effects by interfering with TLR2/4 signaling. In addition, PRG4 has the ability to inhibit activation of cluster of differentiation 44 (CD44), a cell-surface glycoprotein playing an important role in inflammation. Using the controlled cortical impact model of TBI in rats, we showed a rapid and prolonged upregulation of message for TLR2/4 and CD44 in the injured cortex. In the in vitro model of the BBB, recombinant human PRG4 (rhPRG4) crossed the endothelial monolayers through a high-capacity, saturable transport system. In rats sustaining TBI, PRG4 delivery to the brain was enhanced by post-traumatic increase in BBB permeability. rhPRG4 injected intravenously at 1 h post-TBI potently inhibited post-traumatic activation of nuclear factor kappa B and extracellular signal-regulated kinases 1/2, the two major signal transduction pathways associated with TLR2/4 and CD44, and curtailed the post-traumatic influx of monocytes. In addition, PRG4 restored normal BBB function after TBI by preventing the post-traumatic loss of tight junction protein claudin 5 and reduced neuronal death. Our observations provide support for therapeutic strategies targeting TLRs in TBI.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/complicaciones , Encefalitis/tratamiento farmacológico , Proteoglicanos/farmacología , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Muerte Celular/efectos de los fármacos , Encefalitis/etiología , Encefalitis/metabolismo , Encefalitis/patología , Masculino , Modelos Animales , FN-kappa B/metabolismo , Proteoglicanos/uso terapéutico , Ratas , Ratas Long-Evans , Transducción de Señal/efectos de los fármacos
11.
Acta Neuropathol Commun ; 8(1): 4, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31973769

RESUMEN

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.


Asunto(s)
Acetilcisteína/administración & dosificación , Antioxidantes/administración & dosificación , Encéfalo/inmunología , Movimiento Celular/efectos de los fármacos , Líquido Cefalorraquídeo/inmunología , Plexo Coroideo/inmunología , Lipopéptidos/administración & dosificación , Neutrófilos/inmunología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Células Cultivadas , Plexo Coroideo/efectos de los fármacos , Femenino , Mediadores de Inflamación/inmunología , Leucocitos/inmunología , Neutrófilos/efectos de los fármacos , Ratas Sprague-Dawley , Ratas Wistar
12.
PLoS One ; 15(9): e0238301, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32881954

RESUMEN

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.


Asunto(s)
Acuaporina 4/inmunología , Barrera Hematoencefálica/metabolismo , Inmunoglobulina G/farmacología , Neuromielitis Óptica/patología , Permeabilidad/efectos de los fármacos , Animales , Barrera Hematoencefálica/efectos de los fármacos , Células Cultivadas , Quimiocinas/metabolismo , Claudina-5/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Inmunoglobulina G/aislamiento & purificación , Microvasos/citología , Microvasos/metabolismo , Neuromielitis Óptica/metabolismo , Ratas
13.
Fluids Barriers CNS ; 16(1): 41, 2019 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-31856859

RESUMEN

This article highlights the scientific achievements, professional career, and personal interactions of Malcolm B. Segal who passed away in July this year. Born in 1937 in Goodmayes, Essex, UK, Segal rose to the Chairman position in the Division of Physiology at United Medical and Dental School of Guy's and St. Thomas' Hospitals, retiring in 2006 after his long professional career in biomedical science. Being trained in Hugh Davson's laboratory, Segal became one of the pioneers in research on cerebrospinal fluid physiology and the choroid plexus. During the course of his career, Segal himself trained a number of young scientists and collaborated with many colleagues around the world, making long-lasting friendships along the way. In addition to his professional accomplishments as a researcher and educator, Segal was an avid sailor and wine connoisseur, and enjoyed teaching classes on navigation and wine tasting.


Asunto(s)
Líquido Cefalorraquídeo/fisiología , Plexo Coroideo/fisiología , Fisiología/historia , Historia del Siglo XX , Historia del Siglo XXI
14.
Sci Rep ; 9(1): 5998, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30979952

RESUMEN

Many pregnant women and prematurely born infants require medication for clinical conditions including cancer, cardiac defects and psychiatric disorders. In adults drug transfer from blood into brain is mostly restricted by efflux mechanisms (ATP-binding cassette, ABC transporters). These mechanisms have been little studied during brain development. Here expression of eight ABC transporters (abcb1a, abcb1b, abcg2, abcc1, abcc2, abcc3, abcc4, abcc5) and activity of conjugating enzyme glutathione-s-transferase (GST) were measured in livers, brain cortices (blood-brain-barrier) and choroid plexuses (blood-cerebrospinal fluid, CSF, barrier) during postnatal rat development. Controls were compared to animals chronically injected (4 days, 200 mg/kg/day) with known abcb1a inducer diallyl sulfide (DAS). Results reveal both tissue- and age-dependent regulation. In liver abcb1a and abcc3 were up-regulated at all ages. In cortex abcb1a/b, abcg2 and abcc4/abcc5 were up-regulated in adults only, while in choroid plexus abcb1a and abcc2 were up-regulated only at P14. DAS treatment increased GST activity in livers, but not in cortex or choroid plexuses. Immunocytochemistry of ABC transporters at the CSF-brain interface showed that PGP and BCRP predominated in neuroepithelium while MRP2/4/5 were prominent in adult ependyma. These results indicate an age-related capacity of brain barriers to dynamically regulate their defence mechanisms when chronically challenged by xenobiotic compounds.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Compuestos Alílicos/toxicidad , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Sulfuros/toxicidad , Animales , Encéfalo/metabolismo , Glutatión Transferasa/genética , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
15.
Cerebrospinal Fluid Res ; 5: 5, 2008 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-18318891

RESUMEN

BACKGROUND: Prostaglandin E2 (PGE2) is involved in the regulation of synaptic activity and plasticity, and in brain maturation. It is also an important mediator of the central response to inflammatory challenges. The aim of this study was to evaluate the ability of the tissues forming the blood-brain interfaces to act as signal termination sites for PGE2 by metabolic inactivation. METHODS: The specific activity of 15-hydroxyprostaglandin dehydrogenase was measured in homogenates of microvessels, choroid plexuses and cerebral cortex isolated from postnatal and adult rat brain, and compared to the activity measured in peripheral organs which are established signal termination sites for prostaglandins. PGE2 metabolites produced ex vivo by choroid plexuses were identified and quantified by HPLC coupled to radiochemical detection. RESULTS: The data confirmed the absence of metabolic activity in brain parenchyma, and showed that no detectable activity was associated with brain microvessels forming the blood-brain barrier. By contrast, 15-hydroxyprostaglandin dehydrogenase activity was measured in both fourth and lateral ventricle choroid plexuses from 2-day-old rats, albeit at a lower level than in lung or kidney. The activity was barely detectable in adult choroidal tissue. Metabolic profiles indicated that isolated choroid plexus has the ability to metabolize PGE2, mainly into 13,14-dihydro-15-keto-PGE2. In short-term incubations, this metabolite distributed in the tissue rather than in the external medium, suggesting its release in the choroidal stroma. CONCLUSION: The rat choroidal tissue has a significant ability to metabolize PGE2 during early postnatal life. This metabolic activity may participate in signal termination of centrally released PGE2 in the brain, or function as an enzymatic barrier acting to maintain PGE2 homeostasis in CSF during the critical early postnatal period of brain development.

16.
J Infect Dis ; 194(3): 341-9, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16826482

RESUMEN

To probe encephalopathy pathogenesis during toxic shock syndrome (TSS), we investigated the fate of bloodborne TSS toxin-1 (TSST-1) as it moves through the choroid plexus epithelium that forms the main blood-cerebrospinal fluid (CSF) barrier and the effect that TSST-1 has on choroidal barrier properties and on cultured neuronal cell viability. TSST-1 showed a slow, diffusional movement across a cellular model of the blood-CSF barrier but did not compromise the integrity of the barrier. Relevant to the acute symptoms of TSS, a combination of human leukocytes and the toxin induced a decrease in CSF clearance of the pyrogenic prostaglandin E(2) (PGE(2)). The direct effects that TSST-1 had on primary cortical neuron cultures and a neuronal cell line involved elevated caspase 3/7 levels, which correlated with an increase in neuronal cell death. The results of the present study suggest that TSST-1 can affect the brain, by inducing both an intracerebral increase in PGE(2) concentration and caspase-dependent neuronal death, which are possibly relevant to long-term intoxication.


Asunto(s)
Toxinas Bacterianas/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Plexo Coroideo/metabolismo , Enterotoxinas/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Superantígenos/metabolismo , Animales , Apoptosis/fisiología , Toxinas Bacterianas/sangre , Toxinas Bacterianas/líquido cefalorraquídeo , Toxinas Bacterianas/toxicidad , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/patología , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Células Cultivadas , Plexo Coroideo/citología , Dinoprostona/metabolismo , Enterotoxinas/sangre , Enterotoxinas/líquido cefalorraquídeo , Enterotoxinas/toxicidad , Epitelio/inmunología , Epitelio/metabolismo , Humanos , Leucocitos/metabolismo , Neuronas/metabolismo , Neuronas/patología , Síndromes de Neurotoxicidad/microbiología , Ratas , Superantígenos/sangre , Superantígenos/líquido cefalorraquídeo , Superantígenos/toxicidad
17.
J Cereb Blood Flow Metab ; 26(9): 1165-75, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16395287

RESUMEN

The choroid plexuses (CPs) form a protective interface between the blood and the ventricular cerebrospinal fluid (CSF). To probe into the pathways by which CPs provide brain protection, we sought to evaluate the efficiency of glutathione conjugation in this barrier as a mechanism to prevent the entry of blood-borne electrophilic, potentially toxic compounds into the CSF, and we investigated the fate of the resulting metabolites. Rat CPs, as well as human CPs from both fetal and adult brains, displayed high glutathione-S-transferase activities. Using an in vitro model of the blood-CSF barrier consisting of choroidal epithelial cells cultured in a two-chambered device, we showed that glutathione conjugation can efficiently prevent the entry of 1-chloro-2,4-dinitrobenzene (CDNB) into the CSF, a model for electrophilic compounds. The duration of this enzymatic protection was set by the concentration of CDNB to which the epithelium was exposed, and this barrier effect was impaired only on severe epithelial intracellular glutathione and cysteine depletion. The conjugate was excreted from the choroidal cells in a polarized manner, mostly at the blood-facing membrane, via a high-capacity transport process, which is not a rate-limiting step in this detoxification pathway, and which may involve transporters of the ATP-binding cassette c(Abcc) and/or solute carrier 21 (Slc21) families. Supplying the choroidal epithelium at the blood-facing membrane with a therapeutically relevant concentration of N-acetylcysteine sustained this neuroprotective effect. Thus, glutathione conjugation at the CP epithelium coupled with the basolateral efflux of the resulting metabolites form an efficient blood-CSF enzymatic barrier, which can be enhanced by pharmacologically increasing glutathione synthesis within the epithelial cells.


Asunto(s)
Barrera Hematoencefálica/fisiología , Líquido Cefalorraquídeo/fisiología , Glutatión/fisiología , Fármacos Neuroprotectores , Acetilcisteína/metabolismo , Animales , Plexo Coroideo/citología , Plexo Coroideo/efectos de los fármacos , Plexo Coroideo/metabolismo , Cromatografía Líquida de Alta Presión , Cisteína/metabolismo , Dinitroclorobenceno/farmacocinética , Dinitroclorobenceno/toxicidad , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Glutatión Transferasa/metabolismo , Humanos , Irritantes/farmacocinética , Irritantes/toxicidad , Proteínas del Tejido Nervioso/metabolismo , Permeabilidad , ARN/biosíntesis , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Curr Pharm Des ; 22(35): 5463-5476, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27464721

RESUMEN

The blood-brain interfaces restrict the cerebral bioavailability of pharmacological compounds. Various drug delivery strategies have been developed to improve drug penetration into the brain. Most strategies target the microvascular endothelium forming the bloodbrain barrier proper. Targeting the blood-cerebrospinal fluid (CSF) barrier formed by the epithelium of the choroid plexuses in addition to the blood-brain barrier may offer addedvalue for the treatment of central nervous system diseases. For instance, targeting the CSF spaces, adjacent tissue, or the choroid plexuses themselves is of interest for the treatment of neuroinflammatory and infectious diseases, cerebral amyloid angiopathy, selected brain tumors, hydrocephalus or neurohumoral dysregulation. Selected CSF-borne materials seem to reach deep cerebral structures by mechanisms that need to be understood in the context of chronic CSF delivery. Drug delivery through both barriers can reduce CSF sink action towards parenchymal drugs. Finally, targeting the choroid plexus-CSF system can be especially relevant in the context of neonatal and pediatric diseases of the central nervous system. Transcytosis appears the most promising mechanism to target in order to improve drug delivery through brain barriers. The choroid plexus epithelium displays strong vesicular trafficking and secretory activities that deserve to be explored in the context of cerebral drug delivery. Folate transport and exosome release into the CSF, plasma protein transport, and various receptor-mediated endocytosis pathways may prove useful mechanisms to exploit for efficient drug delivery into the CSF. This calls for a clear evaluation of transcytosis mechanisms at the blood-CSF barrier, and a thorough evaluation of CSF drug delivery rates.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Líquido Cefalorraquídeo/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Transporte Biológico/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Sistema Nervioso Central/metabolismo , Líquido Cefalorraquídeo/metabolismo , Humanos
19.
PLoS One ; 11(3): e0150945, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26942913

RESUMEN

An emerging concept of normal brain immune surveillance proposes that recently and moderately activated central memory T lymphocytes enter the central nervous system (CNS) directly into the cerebrospinal fluid (CSF) via the choroid plexus. Within the CSF space, T cells inspect the CNS environment for cognate antigens. This gate of entry into the CNS could also prevail at the initial stage of neuroinflammatory processes. To actually demonstrate T cell migration across the choroidal epithelium forming the blood-CSF barrier, an in vitro model of the rat blood-CSF barrier was established in an "inverse" configuration that enables cell transmigration studies in the basolateral to apical, i.e. blood/stroma to CSF direction. Structural barrier features were evaluated by immunocytochemical analysis of tight junction proteins, functional barrier properties were assessed by measuring the monolayer permeability to sucrose and the active efflux transport of organic anions. The migratory behaviour of activated T cells across the choroidal epithelium was analysed in the presence and absence of chemokines. The migration pathway was examined by confocal microscopy. The inverse rat BCSFB model reproduces the continuous distribution of tight junction proteins at cell margins, the restricted paracellular permeability, and polarized active transport mechanisms, which all contribute to the barrier phenotype in vivo. Using this model, we present experimental evidence of T cell migration across the choroidal epithelium. Cell migration appears to occur via a paracellular route without disrupting the restrictive barrier properties of the epithelial interface. Apical chemokine addition strongly stimulates T cell migration across the choroidal epithelium. The present data provide evidence for the controlled migration of T cells across the blood-CSF barrier into brain. They further indicate that this recruitment route is sensitive to CSF-borne chemokines, extending the relevance of this migration pathway to neuroinflammatory and neuroinfectious disorders which are typified by elevated chemokine levels in CSF.


Asunto(s)
Barrera Hematoencefálica/inmunología , Movimiento Celular , Líquido Cefalorraquídeo/inmunología , Plexo Coroideo/metabolismo , Epitelio/metabolismo , Linfocitos T/citología , Animales , Claudinas/metabolismo , Femenino , Activación de Linfocitos/inmunología , Masculino , Microscopía Confocal , Modelos Biológicos , Ratas , Receptores de Quimiocina/metabolismo , Uniones Estrechas/metabolismo , Migración Transendotelial y Transepitelial
20.
Front Neurosci ; 9: 21, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25698917

RESUMEN

The cerebral microvessel endothelium forming the blood-brain barrier (BBB) and the epithelium of the choroid plexuses forming the blood-CSF barrier (BCSFB) operate as gatekeepers for the central nervous system. Exposure of the vulnerable developing brain to chemical insults can have dramatic consequences for brain maturation and lead to life-long neurological diseases. The ability of blood-brain interfaces to efficiently protect the immature brain is therefore an important pathophysiological issue. This is also key to our understanding of drug entry into the brain of neonatal and pediatric patients. Non-specific paracellular diffusion through barriers is restricted early during development, but other neuroprotective properties of these interfaces differ between the developing and adult brains. This review focuses on the developmental expression and function of various classes of efflux transporters. These include the multispecific transporters of the ATP-binding cassette transporter families ABCB, ABCC, ABCG, the organic anion and cation transporters of the solute carrier families SLC21/SLCO and SLC22, and the peptide transporters of the SLC15 family. These transporters play a key role in preventing brain entry of blood-borne molecules such as drugs, environmental toxicants, and endogenous metabolites, or else in increasing the clearance of potentially harmful organic ions from the brain. The limited data available for laboratory animals and human highlight transporter-specific developmental patterns of expression and function, which differ between blood-brain interfaces. The BCSFB achieves an adult phenotype earlier than BBB. Efflux transporters at the BBB appear to be regulated by various factors subsequently secreted by neural progenitors and astrocytes during development. Their expression is also modulated by oxidative stress, inflammation, and exposure to xenobiotic inducers. A better understanding of these regulatory pathways during development, in particular the signaling pathways triggered by oxidative stress and xenobiotics, may open new opportunities to therapeutic manipulation in view to improve or restore neuroprotective functions of the blood-brain interfaces in the context of perinatal injuries.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA