Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 38(14): 3466-3479, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29507144

RESUMO

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.


Assuntos
Barreira Hematoencefálica/metabolismo , Glutationa Transferase/metabolismo , Glutationa/metabolismo , Animais , Barreira Hematoencefálica/crescimento & desenvolvimento , Plexo Corióideo/crescimento & desenvolvimento , Plexo Corióideo/metabolismo , Epêndima/crescimento & desenvolvimento , Epêndima/metabolismo , Feminino , Radicais Livres/sangue , Radicais Livres/líquido cefalorraquidiano , Glutationa/sangue , Glutationa/líquido cefalorraquidiano , Masculino , Ratos , Ratos Sprague-Dawley
2.
Am J Physiol Cell Physiol ; 315(4): C445-C456, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29949405

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Líquido Cefalorraquidiano/metabolismo , Plexo Corióideo/metabolismo , Peróxido de Hidrogênio/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Células Epiteliais/metabolismo , Feminino , Masculino , Ratos , Ratos Sprague-Dawley
3.
Acta Neuropathol ; 135(3): 337-361, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29368213

RESUMO

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.


Assuntos
Barreira Hematoencefálica/anatomia & histologia , Barreira Hematoencefálica/metabolismo , Ventrículos Cerebrais/anatomia & histologia , Ventrículos Cerebrais/metabolismo , Líquido Cefalorraquidiano/metabolismo , Animais , Barreira Hematoencefálica/patologia , Ventrículos Cerebrais/patologia , Humanos , Neuroproteção/fisiologia
4.
Pharm Res ; 35(4): 84, 2018 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-29516182

RESUMO

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.


Assuntos
Anormalidades Induzidas por Medicamentos/prevenção & controle , Encéfalo/efeitos dos fármacos , Aleitamento Materno , Troca Materno-Fetal/efeitos dos fármacos , Complicações na Gravidez/tratamento farmacológico , Anormalidades Induzidas por Medicamentos/etiologia , Disponibilidade Biológica , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Desenvolvimento Infantil/efeitos dos fármacos , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Humanos , Lactente , Recém-Nascido , Gravidez , Medição de Risco , Distribuição Tecidual
5.
Fluids Barriers CNS ; 21(1): 3, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-38183042

RESUMO

Many neuroscientists use the term Blood-Brain Barrier (BBB) to emphasize restrictiveness, often equating or reducing the notion of BBB properties to tight junction molecules physically sealing cerebral endothelial cells, rather than pointing out the complexity of this biological interface with respect to its selectivity and variety of exchange between the general blood circulation and the central nervous tissue. Several authors in the field find it unfortunate that the exquisitely dynamic interfaces between blood and brain continue to be viewed primarily as obstructive barriers to transport. Although the term blood-brain interface is an excellent descriptor that does not convey the idea of a barrier, it is important and preferable for the spreading of an idea beyond specialist communities to try to appeal to well-chosen metaphors. Recent evidence reviewed here indicates that blood-brain interfaces are more than selective semi-permeable membranes in that they display many dynamic processes and complex mechanisms for communication. They are thus more like 'geopolitical borders'. Furthermore, some authors working on blood-brain interface-relevant issues have started to use the word border, for example in border-associated macrophages. Therefore, we suggest adopting the term Blood-Brain Border to better communicate the flexibility of and movement across blood-brain interfaces.


Assuntos
Barreira Hematoencefálica , Sistema Cardiovascular , Células Endoteliais , Encéfalo , Junções Íntimas
6.
Neuropathology ; 33(1): 17-29, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22537279

RESUMO

Circumventricular organs (CVOs) are specialized ventricular structures around the third and fourth ventricles of the brain. In humans, these structures are present during the fetal period and some become vestigial after birth. Some of these organs, such as the pineal gland (PG), subcommissural organ (SCO), and organum vasculosum of the lamina terminalis, might be the sites of origin of periventricular tumors, notably pineal parenchymal tumors, papillary tumor of the pineal region and chordoid glioma. In contrast to the situation in humans, CVOs are present in the adult rat and can be dissected by laser capture microdissection (LCM). In this study, we used LCM and microarrays to analyze the transcriptomes of three CVOs, the SCO, the subfornical organ (SFO), and the PG and the third ventricle ependyma in the adult rat, in order to better characterize these organs at the molecular level. Several genes were expressed only, or mainly, in one of these structures, for example, Erbb2 and Col11a1 in the ependyma, Epcam and Claudin-3 (CLDN3) in the SCO, Ren1 and Slc22a3 in the SFO and Tph, Aanat and Asmt in the PG. The expression of these genes in periventricular tumors should be examined as evidence for a possible origin from the CVOs. Furthermore, we performed an immunohistochemical study of CLDN3, a membrane protein involved in forming cellular tight junctions and found that CLDN3 expression was restricted to the apical pole of ependymocytes in the SCO. This microarray study provides new evidence regarding the possible origin of some rare periventricular tumors.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias do Ventrículo Cerebral/metabolismo , Glândula Pineal/metabolismo , Órgão Subcomissural/metabolismo , Órgão Subfornical/metabolismo , Animais , Ventrículos Cerebrais/metabolismo , Epêndima/metabolismo , Microdissecção e Captura a Laser , Masculino , Ratos , Ratos Sprague-Dawley , Transcriptoma
7.
J Neuroinflammation ; 9: 187, 2012 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-22870891

RESUMO

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.


Assuntos
Encefalomielite Autoimune Experimental/líquido cefalorraquidiano , Encefalomielite Autoimune Experimental/patologia , Leucócitos/patologia , Prosencéfalo/patologia , Sequência de Aminoácidos , Animais , Biomarcadores/líquido cefalorraquidiano , Encefalomielite Autoimune Experimental/imunologia , Feminino , Cobaias , Inflamação/líquido cefalorraquidiano , Inflamação/imunologia , Inflamação/patologia , Leucócitos/imunologia , Leucócitos/metabolismo , Mesencéfalo/imunologia , Mesencéfalo/metabolismo , Mesencéfalo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Prosencéfalo/metabolismo , Ratos , Transdução de Sinais/imunologia
8.
Histochem Cell Biol ; 138(6): 861-79, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22886143

RESUMO

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.


Assuntos
Barreira Hematoencefálica/metabolismo , Claudinas/análise , Claudinas/genética , Perfilação da Expressão Gênica , Animais , Western Blotting , Plexo Corióideo/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Junções Íntimas/metabolismo
9.
Neurotoxicology ; 91: 100-118, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35436567

RESUMO

Endocrine disrupting chemicals (EDCs) act on peripheral endocrine organs or interfere with general endocrine pathways. Several EDCs alter the central regulation of neuroendocrine pathways, and affect neurological functions, and as such can be classified as neurotoxic molecules. Environmental pollutants classified as EDCs and affecting the central nervous system include perfluoroalcanes, parabens, phthalates, organotins, bisphenols, benzophenones, polychlorinated biphenyls, and dioxins. In this review we provide a brief description of these families of EDCs. We report and compare the EDC concentrations measured in the brain of humans and wild animals naturally exposed to these molecules, as well as in the brain of laboratory animals experimentally exposed to EDCs. The importance of using sophisticated analytical tools to detect EDCs in the brain is pointed out. The ability of blood-brain interfaces to reduce the brain exposition to EDCs in adult and during development is discussed in relation with the specific morphological, transport and metabolic properties of these cellular layers. Finally, we review the evidence that the neuroprotective functions of blood-brain interfaces can be altered by EDCs, a process that may participate to the central toxic action of these molecules. Overall this analysis points to the implication of blood-brain interfaces in setting the extent of central EDCs toxicity, although most evidence is indirect. Therefore, more specific blood-brain interface-oriented studies are called for in this field of EDC neurotoxicology.


Assuntos
Disruptores Endócrinos , Poluentes Ambientais , Bifenilos Policlorados , Adulto , Animais , Barreira Hematoencefálica , Disruptores Endócrinos/toxicidade , Poluentes Ambientais/toxicidade , Humanos , Sistemas Neurossecretores , Bifenilos Policlorados/toxicidade
10.
Fluids Barriers CNS ; 19(1): 47, 2022 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-35672829

RESUMO

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.


Assuntos
Barreira Hematoencefálica , Icterícia Neonatal , Animais , Bilirrubina/metabolismo , Barreira Hematoencefálica/metabolismo , Líquido Cefalorraquidiano/metabolismo , Plexo Corióideo/metabolismo , Citocinas/metabolismo , Humanos , Hiperbilirrubinemia/metabolismo , Recém-Nascido , Icterícia Neonatal/metabolismo , Ratos , Ratos Gunn , Sacarose
11.
J Neurochem ; 117(4): 747-56, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21395586

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Manganês/líquido cefalorraquidiano , Manganês/metabolismo , Animais , Transporte Biológico Ativo , Barreira Hematoencefálica , Cálcio/metabolismo , Cálcio/farmacologia , Permeabilidade da Membrana Celular , Células Cultivadas , Plexo Corióideo/metabolismo , Cisteína/metabolismo , Dinoprostona/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Glutationa/metabolismo , Indicadores e Reagentes , Masculino , Manganês/sangue , Metais/metabolismo , Ratos , Ratos Sprague-Dawley , Sacarose/metabolismo , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
12.
J Neurotrauma ; 38(4): 385-398, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-32940130

RESUMO

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.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Lesões Encefálicas Traumáticas/complicações , Encefalite/tratamento farmacológico , Proteoglicanas/farmacologia , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Morte Celular/efeitos dos fármacos , Encefalite/etiologia , Encefalite/metabolismo , Encefalite/patologia , Masculino , Modelos Animais , NF-kappa B/metabolismo , Proteoglicanas/uso terapêutico , Ratos , Ratos Long-Evans , Transdução de Sinais/efeitos dos fármacos
13.
PLoS One ; 15(9): e0238301, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32881954

RESUMO

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.


Assuntos
Aquaporina 4/imunologia , Barreira Hematoencefálica/metabolismo , Imunoglobulina G/farmacologia , Neuromielite Óptica/patologia , Permeabilidade/efeitos dos fármacos , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Células Cultivadas , Quimiocinas/metabolismo , Claudina-5/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Imunoglobulina G/isolamento & purificação , Microvasos/citologia , Microvasos/metabolismo , Neuromielite Óptica/metabolismo , Ratos
14.
Acta Neuropathol Commun ; 8(1): 4, 2020 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-31973769

RESUMO

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.


Assuntos
Acetilcisteína/administração & dosagem , Antioxidantes/administração & dosagem , Encéfalo/imunologia , Movimento Celular/efeitos dos fármacos , Líquido Cefalorraquidiano/imunologia , Plexo Corióideo/imunologia , Lipopeptídeos/administração & dosagem , Neutrófilos/imunologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Células Cultivadas , Plexo Corióideo/efeitos dos fármacos , Feminino , Mediadores da Inflamação/imunologia , Leucócitos/imunologia , Neutrófilos/efeitos dos fármacos , Ratos Sprague-Dawley , Ratos Wistar
15.
Fluids Barriers CNS ; 16(1): 41, 2019 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-31856859

RESUMO

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.


Assuntos
Líquido Cefalorraquidiano/fisiologia , Plexo Corióideo/fisiologia , Fisiologia/história , História do Século XX , História do Século XXI
16.
Front Mol Neurosci ; 12: 34, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30809124

RESUMO

The choroid plexuses (ChPs) perform indispensable functions for the development, maintenance and functioning of the brain. Although they have gained considerable interest in the last years, their involvement in brain disorders is still largely unknown, notably because their deep location inside the brain hampers non-invasive investigations. Imaging tools have become instrumental to the diagnosis and pathophysiological study of neurological and neuropsychiatric diseases. This review summarizes the knowledge that has been gathered from the clinical imaging of ChPs in health and brain disorders not related to ChP pathologies. Results are discussed in the light of pre-clinical imaging studies. As seen in this review, to date, most clinical imaging studies of ChPs have used disease-free human subjects to demonstrate the value of different imaging biomarkers (ChP size, perfusion/permeability, glucose metabolism, inflammation), sometimes combined with the study of normal aging. Although very few studies have actually tested the value of ChP imaging biomarkers in patients with brain disorders, these pioneer studies identified ChP changes that are promising data for a better understanding and follow-up of diseases such as schizophrenia, epilepsy and Alzheimer's disease. Imaging of immune cell trafficking at the ChPs has remained limited to pre-clinical studies so far but has the potential to be translated in patients for example using MRI coupled with the injection of iron oxide nanoparticles. Future investigations should aim at confirming and extending these findings and at developing translational molecular imaging tools for bridging the gap between basic molecular and cellular neuroscience and clinical research.

17.
Sci Rep ; 9(1): 5998, 2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30979952

RESUMO

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.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Compostos Alílicos/toxicidade , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Sulfetos/toxicidade , Animais , Encéfalo/metabolismo , Glutationa Transferase/genética , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
18.
Cerebrospinal Fluid Res ; 5: 5, 2008 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-18318891

RESUMO

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.

20.
J Infect Dis ; 194(3): 341-9, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16826482

RESUMO

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.


Assuntos
Toxinas Bacterianas/metabolismo , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Plexo Corióideo/metabolismo , Enterotoxinas/metabolismo , Síndromes Neurotóxicas/metabolismo , Superantígenos/metabolismo , Animais , Apoptose/fisiologia , Toxinas Bacterianas/sangue , Toxinas Bacterianas/líquido cefalorraquidiano , Toxinas Bacterianas/toxicidade , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/patologia , Caspase 3/metabolismo , Caspase 7/metabolismo , Células Cultivadas , Plexo Corióideo/citologia , Dinoprostona/metabolismo , Enterotoxinas/sangue , Enterotoxinas/líquido cefalorraquidiano , Enterotoxinas/toxicidade , Epitélio/imunologia , Epitélio/metabolismo , Humanos , Leucócitos/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Síndromes Neurotóxicas/microbiologia , Ratos , Superantígenos/sangue , Superantígenos/líquido cefalorraquidiano , Superantígenos/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA