Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Fluids Barriers CNS ; 21(1): 51, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858667

RESUMO

Oedema occurs when higher than normal amounts of solutes and water accumulate in tissues. In brain parenchymal tissue, vasogenic oedema arises from changes in blood-brain barrier permeability, e.g. in peritumoral oedema. Cytotoxic oedema arises from excess accumulation of solutes within cells, e.g. ischaemic oedema following stroke. This type of oedema is initiated when blood flow in the affected core region falls sufficiently to deprive brain cells of the ATP needed to maintain ion gradients. As a consequence, there is: depolarization of neurons; neural uptake of Na+ and Cl- and loss of K+; neuronal swelling; astrocytic uptake of Na+, K+ and anions; swelling of astrocytes; and reduction in ISF volume by fluid uptake into neurons and astrocytes. There is increased parenchymal solute content due to metabolic osmolyte production and solute influx from CSF and blood. The greatly increased [K+]isf triggers spreading depolarizations into the surrounding penumbra increasing metabolic load leading to increased size of the ischaemic core. Water enters the parenchyma primarily from blood, some passing into astrocyte endfeet via AQP4. In the medium term, e.g. after three hours, NaCl permeability and swelling rate increase with partial opening of tight junctions between blood-brain barrier endothelial cells and opening of SUR1-TPRM4 channels. Swelling is then driven by a Donnan-like effect. Longer term, there is gross failure of the blood-brain barrier. Oedema resolution is slower than its formation. Fluids without colloid, e.g. infused mock CSF, can be reabsorbed across the blood-brain barrier by a Starling-like mechanism whereas infused serum with its colloids must be removed by even slower extravascular means. Large scale oedema can increase intracranial pressure (ICP) sufficiently to cause fatal brain herniation. The potentially lethal increase in ICP can be avoided by craniectomy or by aspiration of the osmotically active infarcted region. However, the only satisfactory treatment resulting in retention of function is restoration of blood flow, providing this can be achieved relatively quickly. One important objective of current research is to find treatments that increase the time during which reperfusion is successful. Questions still to be resolved are discussed.


Assuntos
Edema Encefálico , Encéfalo , Humanos , Edema Encefálico/fisiopatologia , Edema Encefálico/metabolismo , Edema Encefálico/etiologia , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/fisiopatologia , Isquemia Encefálica/fisiopatologia , Isquemia Encefálica/metabolismo
2.
Fluids Barriers CNS ; 19(1): 9, 2022 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-35115036

RESUMO

The glymphatic hypothesis proposes a mechanism for extravascular transport into and out of the brain of hydrophilic solutes unable to cross the blood-brain barrier. It suggests that there is a circulation of fluid carrying solutes inwards via periarterial routes, through the interstitium and outwards via perivenous routes. This review critically analyses the evidence surrounding the mechanisms involved in each of these stages. There is good evidence that both influx and efflux of solutes occur along periarterial routes but no evidence that the principal route of outflow is perivenous. Furthermore, periarterial inflow of fluid is unlikely to be adequate to provide the outflow that would be needed to account for solute efflux. A tenet of the hypothesis is that flow sweeps solutes through the parenchyma. However, the velocity of any possible circulatory flow within the interstitium is too small compared to diffusion to provide effective solute movement. By comparison the earlier classical hypothesis describing extravascular transport proposed fluid entry into the parenchyma across the blood-brain barrier, solute movements within the parenchyma by diffusion, and solute efflux partly by diffusion near brain surfaces and partly carried by flow along "preferred routes" including perivascular spaces, white matter tracts and subependymal spaces. It did not suggest fluid entry via periarterial routes. Evidence is still incomplete concerning the routes and fate of solutes leaving the brain. A large proportion of the solutes eliminated from the parenchyma go to lymph nodes before reaching blood but the proportions delivered directly to lymph or indirectly via CSF which then enters lymph are as yet unclear. In addition, still not understood is why and how the absence of AQP4 which is normally highly expressed on glial endfeet lining periarterial and perivenous routes reduces rates of solute elimination from the parenchyma and of solute delivery to it from remote sites of injection. Neither the glymphatic hypothesis nor the earlier classical hypothesis adequately explain how solutes and fluid move into, through and out of the brain parenchyma. Features of a more complete description are discussed. All aspects of extravascular transport require further study.


Assuntos
Transporte Biológico , Sistema Glinfático/fisiologia , Animais , Humanos
3.
Fluids Barriers CNS ; 16(1): 24, 2019 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-31299992

RESUMO

Solutes can enter and leave gray matter in the brain by perivascular routes. The glymphatic hypothesis supposes that these movements are a consequence of inward flow along periarterial spaces and an equal outward flow along perivenous spaces. The flow through the parenchyma between periarterial and perivenous spaces is the same as the inflow and the outflow. Ray et al. (Fluids Barriers CNS 16:6, 2019) have investigated how this flow could interact with diffusion using numerical simulations of real-time iontophoresis experiments that monitor the concentrations of tetramethylammonium ions (TMA+) injected into the parenchyma via iontophoresis. For this purpose they have devised a description of the parenchyma incorporating perivascular spaces. Their simulations show that superficial flow velocities of about 50 µm min-1 are needed to produce changes in TMA+ fluxes comparable to those accounted for by diffusion. In the glymphatic hypothesis the proposed flow through the parenchyma can be estimated from the clearance of solutes that are present in the perivenous outflow at the same concentration as in the interstitial fluid of the parenchyma. Reported clearances are approximately 1 µL min-1 g-1. This flow can be converted to a superficial flow velocity using the area available for the flow, which can be estimated using Ray et al.'s description of the tissue as 40 cm2 g-1. The best available estimate of the flow velocity is thus 0.25 µm min-1 which is 200 times smaller than the flow that produces effects comparable to diffusion for TMA+. Thus it follows in Ray et al.'s description of the parenchyma that diffusion rather than flow accounts for TMA+ movements. Because the diffusion constant depends only weakly on molecular weight the same is expected to apply even for solutes somewhat larger than serum albumin.


Assuntos
Encéfalo/metabolismo , Líquido Extracelular/metabolismo , Espaço Extracelular/metabolismo , Substância Cinzenta/metabolismo , Transporte Biológico/fisiologia , Difusão , Humanos
4.
Handb Exp Pharmacol ; 253: 385-423, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-28744585

RESUMO

Mechanisms for elimination of metabolites from ISF include metabolism, blood-brain barrier transport and non-selective, perivascular efflux, this last being assessed by measuring the clearance of markers like inulin. Clearance describes elimination. Clearance of a metabolite generated within the brain is determined as its elimination rate divided by its concentration in interstitial fluid (ISF). However, the more frequently measured parameter is the rate constant for elimination determined as elimination rate divided by amount present, which thus depends on both the elimination processes and the distribution of the metabolite in the brain. The relative importance of the various elimination mechanisms depends on the particular metabolite. Little is known about the effects of sleep on clearance via metabolism or blood-brain barrier transport, but studies with inulin in mice comparing perivascular effluxes during sleep and wakefulness reveal a 4.2-fold increase in clearance. Amongst the important brain metabolites considered, CO2 is eliminated so rapidly across the blood-brain barrier that clearance is blood flow limited and elimination quickly balances production. Glutamate is removed from ISF primarily by uptake into astrocytes and conversion to glutamine, but also by transport across the blood-brain barrier. Both lactate and amyloid-ß are eliminated by metabolism, blood-brain barrier transport and perivascular efflux and both show decreased production, decreased ISF concentration and increased perivascular clearance during sleep. Taken altogether available data indicate that sleep increases perivascular and non-perivascular clearances for amyloid-ß which reduces its concentration and may have long-term consequences for the formation of plaques and cerebral arterial deposits.


Assuntos
Barreira Hematoencefálica , Encéfalo/metabolismo , Vigília , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Camundongos , Sono
5.
Fluids Barriers CNS ; 15(1): 30, 2018 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-30340614

RESUMO

This review considers efflux of substances from brain parenchyma quantified as values of clearances (CL, stated in µL g-1 min-1). Total clearance of a substance is the sum of clearance values for all available routes including perivascular pathways and the blood-brain barrier. Perivascular efflux contributes to the clearance of all water-soluble substances. Substances leaving via the perivascular routes may enter cerebrospinal fluid (CSF) or lymph. These routes are also involved in entry to the parenchyma from CSF. However, evidence demonstrating net fluid flow inwards along arteries and then outwards along veins (the glymphatic hypothesis) is still lacking. CLperivascular, that via perivascular routes, has been measured by following the fate of exogenously applied labelled tracer amounts of sucrose, inulin or serum albumin, which are not metabolized or eliminated across the blood-brain barrier. With these substances values of total CL ≅ 1 have been measured. Substances that are eliminated at least partly by other routes, i.e. across the blood-brain barrier, have higher total CL values. Substances crossing the blood-brain barrier may do so by passive, non-specific means with CLblood-brain barrier values ranging from < 0.01 for inulin to > 1000 for water and CO2. CLblood-brain barrier values for many small solutes are predictable from their oil/water partition and molecular weight. Transporters specific for glucose, lactate and many polar substrates facilitate efflux across the blood-brain barrier producing CLblood-brain barrier values > 50. The principal route for movement of Na+ and Cl- ions across the blood-brain barrier is probably paracellular through tight junctions between the brain endothelial cells producing CLblood-brain barrier values ~ 1. There are large fluxes of amino acids into and out of the brain across the blood-brain barrier but only small net fluxes have been observed suggesting substantial reuse of essential amino acids and α-ketoacids within the brain. Amyloid-ß efflux, which is measurably faster than efflux of inulin, is primarily across the blood-brain barrier. Amyloid-ß also leaves the brain parenchyma via perivascular efflux and this may be important as the route by which amyloid-ß reaches arterial walls resulting in cerebral amyloid angiopathy.


Assuntos
Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Sistema Glinfático/metabolismo , Tecido Parenquimatoso/metabolismo , Animais , Encéfalo/irrigação sanguínea , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Modelos Neurológicos , Tecido Parenquimatoso/irrigação sanguínea
7.
Fluids Barriers CNS ; 13(1): 19, 2016 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-27799072

RESUMO

The two major interfaces separating brain and blood have different primary roles. The choroid plexuses secrete cerebrospinal fluid into the ventricles, accounting for most net fluid entry to the brain. Aquaporin, AQP1, allows water transfer across the apical surface of the choroid epithelium; another protein, perhaps GLUT1, is important on the basolateral surface. Fluid secretion is driven by apical Na+-pumps. K+ secretion occurs via net paracellular influx through relatively leaky tight junctions partially offset by transcellular efflux. The blood-brain barrier lining brain microvasculature, allows passage of O2, CO2, and glucose as required for brain cell metabolism. Because of high resistance tight junctions between microvascular endothelial cells transport of most polar solutes is greatly restricted. Because solute permeability is low, hydrostatic pressure differences cannot account for net fluid movement; however, water permeability is sufficient for fluid secretion with water following net solute transport. The endothelial cells have ion transporters that, if appropriately arranged, could support fluid secretion. Evidence favours a rate smaller than, but not much smaller than, that of the choroid plexuses. At the blood-brain barrier Na+ tracer influx into the brain substantially exceeds any possible net flux. The tracer flux may occur primarily by a paracellular route. The blood-brain barrier is the most important interface for maintaining interstitial fluid (ISF) K+ concentration within tight limits. This is most likely because Na+-pumps vary the rate at which K+ is transported out of ISF in response to small changes in K+ concentration. There is also evidence for functional regulation of K+ transporters with chronic changes in plasma concentration. The blood-brain barrier is also important in regulating HCO3- and pH in ISF: the principles of this regulation are reviewed. Whether the rate of blood-brain barrier HCO3- transport is slow or fast is discussed critically: a slow transport rate comparable to those of other ions is favoured. In metabolic acidosis and alkalosis variations in HCO3- concentration and pH are much smaller in ISF than in plasma whereas in respiratory acidosis variations in pHISF and pHplasma are similar. The key similarities and differences of the two interfaces are summarized.


Assuntos
Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Líquido Cefalorraquidiano/metabolismo , Animais , Humanos , Transporte de Íons/fisiologia
8.
Pflugers Arch ; 466(5): 887-901, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24022703

RESUMO

Ions and water transported across the endothelium lining the blood­brain barrier contribute to the fluid secreted into the brain and are important in maintaining appropriate volume and ionic composition of brain interstitial fluid. Changes in this secretion process may occur after stroke. The present study identifies at transcript and protein level ion transporters involved in the movement of key ions and examines how levels of certain of these alter following oxidative stress. Immunohistochemistry provides evidence for Cl−/HCO3− exchanger, AE2, and Na+, HCO3− cotransporters, NBCe1 and NBCn1, on brain microvessels. mRNA analysis by RT-PCR reveals expression of these transporters in cultured rat brain microvascular endothelial cells (both primary and immortalized GPNT cells) and also Na+/H+ exchangers, NHE1 (primary and immortalized) and NHE2 (primary cells only). Knock-down using siRNA in immortalized GPNT cells identifies AE2 as responsible for much of the Cl−/HCO3− exchange following extracellular chloride removal and NHE1 as the transporter that accounts for most of the Na+/H+ exchange following intracellular acidification. Transcript levels of both AE2 and NHE1 are increased following hypoxia/reoxygenation. Further work is now required to determine the localization of the bicarbonate transporters to luminal or abluminal membranes of the endothelial cells as well as to identify and localize additional transport mechanisms that must exist for K+ and Cl−.


Assuntos
Barreira Hematoencefálica/metabolismo , Antiportadores de Cloreto-Bicarbonato/metabolismo , Células Endoteliais/metabolismo , Líquido Extracelular/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Barreira Hematoencefálica/citologia , Permeabilidade Capilar , Linhagem Celular , Células Cultivadas , Antiportadores de Cloreto-Bicarbonato/genética , Transporte de Íons , Microvasos/citologia , Microvasos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Simportadores de Sódio-Bicarbonato/genética , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética
9.
Fluids Barriers CNS ; 11(1): 26, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25678956

RESUMO

Interstitial fluid (ISF) surrounds the parenchymal cells of the brain and spinal cord while cerebrospinal fluid (CSF) fills the larger spaces within and around the CNS. Regulation of the composition and volume of these fluids is important for effective functioning of brain cells and is achieved by barriers that prevent free exchange between CNS and blood and by mechanisms that secrete fluid of controlled composition into the brain and distribute and reabsorb it. Structures associated with this regular fluid turnover include the choroid plexuses, brain capillaries comprising the blood-brain barrier, arachnoid villi and perineural spaces penetrating the cribriform plate. ISF flow, estimated from rates of removal of markers from the brain, has been thought to reflect rates of fluid secretion across the blood-brain barrier, although this has been questioned because measurements were made under barbiturate anaesthesia possibly affecting secretion and flow and because CSF influx to the parenchyma via perivascular routes may deliver fluid independently of blood-brain barrier secretion. Fluid secretion at the blood-brain barrier is provided by specific transporters that generate solute fluxes so creating osmotic gradients that force water to follow. Any flow due to hydrostatic pressures driving water across the barrier soon ceases unless accompanied by solute transport because water movements modify solute concentrations. CSF is thought to be derived primarily from secretion by the choroid plexuses. Flow rates measured using phase contrast magnetic resonance imaging reveal CSF movements to be more rapid and variable than previously supposed, even implying that under some circumstances net flow through the cerebral aqueduct may be reversed with net flow into the third and lateral ventricles. Such reversed flow requires there to be alternative sites for both generation and removal of CSF. Fluorescent tracer analysis has shown that fluid flow can occur from CSF into parenchyma along periarterial spaces. Whether this represents net fluid flow and whether there is subsequent flow through the interstitium and net flow out of the cortex via perivenous routes, described as glymphatic circulation, remains to be established. Modern techniques have revealed complex fluid movements within the brain. This review provides a critical evaluation of the data.

10.
Biochem J ; 448(3): 389-400, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22950671

RESUMO

Glutathione export from uninfected human erythrocytes was compared with that from cells infected with the malaria parasite Plasmodium falciparum using two separate methods that distinguish between oxidized (GSSG) and reduced (GSH) glutathione. One involved enzymatic recycling with or without thiol-masking; the other involved rapid derivatization followed by HPLC. Glutathione efflux from uninfected erythrocytes under physiological conditions occurred predominantly as GSH. On exposure of the cells to oxidative challenge, efflux of GSSG exceeded that of GSH. Efflux of both species was blocked by MK571, an inhibitor of mammalian multidrug-resistance proteins. Glutathione efflux from parasitized erythrocytes was substantially greater than that from uninfected erythrocytes. Under physiological conditions, the exported species was GSH, whereas under energy-depleted conditions, GSSG efflux occurred. Glutathione export from parasitized cells was inhibited partially by MK571 and more so by furosemide, an inhibitor of the 'new permeability pathways' induced by the parasite in the host erythrocyte membrane. Efflux from isolated parasites occurred as GSH. On exposure to oxidative challenge, this GSH efflux decreased, but no GSSG export was detected. These results are consistent with the view that the parasite supplies its host erythrocyte with GSH, much of which is exported from the infected cell via parasite-induced pathways.


Assuntos
Eritrócitos/metabolismo , Eritrócitos/parasitologia , Glutationa/sangue , Plasmodium falciparum/metabolismo , Animais , Transporte Biológico Ativo/efeitos dos fármacos , Células Cultivadas , Eritrócitos/efeitos dos fármacos , Humanos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/antagonistas & inibidores , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Oxirredução/efeitos dos fármacos , Plasmodium falciparum/isolamento & purificação , Propionatos/farmacologia , Quinolinas/farmacologia
11.
Brain Res ; 1418: 1-11, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-21920506

RESUMO

ABC (ATP Binding Cassette) efflux transporters at the blood-brain barrier, P-glycoprotein (ABCB1), multidrug resistance associated protein 4 (ABCC4) and breast cancer resistance protein (ABCG2), are important for protecting the brain from circulating xenobiotics. Their expression is regulated by signals from surrounding brain tissue that may alter in CNS pathologies. Differences have been reported in transporter expression on brain vasculature of Alzheimer's subjects where raised levels of ß-amyloid (Aß) occur. The present study examines in vitro the effects of Aß using immortalised brain endothelial cells (hCMEC/D3). Significantly lower expression of ABCB1 but not ABCC4 or ABCG2 was found following exposure to Aß(1-42) peptide but not its scrambled equivalent. This was evident at both protein and transcript level and was reflected in lower transcriptional activity of the ABCB1 promoter as judged from the luciferase reporter gene assay and in decreases in ABCB1-mediated efflux of rhodamine 123. Aß exposure also affected Wnt/ß-catenin signalling, decreasing levels of ß-catenin protein, reducing activation of TOPFLASH and increasing transcript levels of endogenous inhibitor, Dkk-1. Application of Wnt3a reversed the Aß-induced changes to ABCB1 protein. These results suggest that Aß may impair Wnt/ß-catenin signalling at the blood-brain barrier but that activation of this pathway may restore ABCB1.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Peptídeos beta-Amiloides/farmacologia , Encéfalo/citologia , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Transporte Biológico/efeitos dos fármacos , Linhagem Celular Transformada , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , RNA Mensageiro/metabolismo , Rodamina 123/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sais de Tetrazólio , Tiazóis , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
12.
Cell Mol Neurobiol ; 31(7): 1103-11, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21618049

RESUMO

Ischemia-reperfusion leads to increased levels at the blood-brain barrier of the multidrug efflux transporter, P-glycoprotein that provides protection to the brain by limiting access of unwanted substances. This is coincident with the production of nitric oxide. This present study using immortalized rat brain endothelial cells (GPNTs) examines whether following hypoxia-reoxygenation, nitric oxide contributes to the alterations in P-glycoprotein levels. After 6 h of hypoxia, both nitric oxide and reactive oxygen species, detected intracellularly using fluorescent monitoring dyes, were produced in the subsequent reoxygenation phase coincident with increased P-glycoprotein. The evidence that nitric oxide can directly affect P-glycoprotein expression was sought by applying S-nitroso-N-acetyl-DL: -penicillamine that as shown increased the nitric oxide generation. Sodium nitroprusside, though more effective at increasing P-glycoprotein expression, appeared to produce different reactive species. Real time RT-PCR analysis revealed the predominant form of nitric oxide synthase in these cells to be endothelial, inhibition of which partially prevented the increase in P-glycoprotein during reoxygenation. These data indicate that the production of nitric oxide by endothelial nitric oxide synthase during reoxygenation can influence P-glycoprotein expression in cells of the blood-rat brain barrier, highlighting another route by which nitric oxide may protect the brain.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Encéfalo/irrigação sanguínea , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Hipóxia/metabolismo , Óxido Nítrico/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Linhagem Celular , Células Endoteliais/citologia , Doadores de Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Oxigênio/metabolismo , Ratos , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo
13.
Brain Res ; 1358: 228-38, 2010 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-20727860

RESUMO

Multidrug efflux transporters of the ATP-Binding cassette (ABC) family, P-glycoprotein (Pgp), multidrug-resistance associated protein 4 (MRP4) and breast cancer resistance protein (BCRP), located on endothelial cells lining brain vasculature play important roles in limiting movement of substances into and enhancing their efflux from the brain. Signals from the surrounding brain normally maintain such barrier function but these may become altered in CNS pathologies such as Alzheimer's disease (AD). Previous studies have reported decreases in the glucose transporter, Glut-1, in brain vasculature of AD patients. The present study investigates the status of the multidrug efflux transporters. Sections of frozen brain from hippocampal region obtained from male AD and age-matched non-demented cases were examined for amyloid plaques and Dkk-1 expression and subjected to dual fluorescence immunochemical staining using antibodies against Pgp, BCRP or MRP4 and von Willebrand factor. Protein expression of each transporter was assessed using confocal microscopy, quantifying peak fluorescence values of cross sectional profiles across brain microvessels. Results in brain microvessels revealed expression of Pgp protein to be significantly lower in hippocampal vessels of patients with AD compared to normal individuals whereas that of MRP4 or BCRP protein was not. By contrast, analysis of the sections at protein level via Western blotting or at transcript level by qRT-PCR did not reveal significantly lower expression for either Pgp or BCRP. Such analysis did however reveal higher than normal expression in the AD brains of MRP4, probably due to gliosis, MRP4 being present also in glial cells.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Doença de Alzheimer/patologia , Encéfalo/patologia , Células Endoteliais/metabolismo , Microvasos/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Idoso , Idoso de 80 Anos ou mais , Amiloide/metabolismo , Estudos de Casos e Controles , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Placa Amiloide/patologia , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo
14.
J Neurochem ; 111(1): 132-41, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19656260

RESUMO

Levels of multidrug efflux transporter P-glycoprotein (P-gp) on endothelial cells lining brain blood vessels are important for limiting access of many compounds to the brain. In vivo studies have indicated that ischaemia-reperfusion that generates reactive oxygen species also increases P-gp levels in brain endothelial cells. To investigate possible mechanisms, in vitro studies were performed on immortalised (GPNT) and primary rat brain endothelial cells. Exposure to hydrogen peroxide (200 microM) resulted in intracellular oxidative stress as detected from higher levels of dichlorofluorescein fluorescence and raised levels of P-gp protein, mdr1a and mdr1b transcripts and, in GPNT cells, increased mdr1a and mdr1b promoter activity. The P-gp protein increases were abolished by pre-treatment with polyethylene glycol-catalase and were curtailed by co-culture with primary rat astrocytes. Exposure of GPNT cells to 6 h hypoxia followed by 24 h reoxygenation produced less intracellular oxidative stress as judged from smaller increments in dichlorofluorescein fluorescence but still resulted in raised levels of P-gp protein, an effect partially abolished by pre-treatment with polyethylene glycol-catalase. However, transcript levels and promoter activities were not significantly increased. These data suggest that hydrogen peroxide contributes to P-gp up-regulation following hypoxia-reoxygenation but the underlying mechanisms of its actions differ from those occurring after direct hydrogen peroxide application.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Células Endoteliais/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Hipóxia/patologia , Oxidantes/farmacologia , Oxigênio/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/farmacologia , Animais , Astrócitos , Encéfalo/citologia , Catalase/farmacologia , Linhagem Celular Transformada , Células Cultivadas , Técnicas de Cocultura/métodos , Células Endoteliais/metabolismo , Fluoresceínas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hipóxia/fisiopatologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transfecção/métodos , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
15.
Brain Res ; 1276: 1-10, 2009 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-19393634

RESUMO

Multidrug efflux transporters protect cells in the brain from potentially harmful substances but also from therapeutically useful drugs. Thus any condition that causes changes in their expression is of some importance with regard to drug access. In this study, changes in efflux transporter expression are investigated in mice containing a mutant constitutively active glycogen synthase kinase-3 (GSK-3beta) transgene, driven by the Thy-1 promoter so limiting its localization predominantly to neurons and some glial cells. As expected, decreases in beta-catenin were evident via Western blot analyses of cortical homogenates prepared from brains of these transgenic mice. As assessed by real time qRT-PCR, decreased transcript levels of the mdr1b isoform of P-glycoprotein, Mrp1 and Mrp4, (transporters associated with neurons and/or glial cells) were observed in the cortex but not the subventricular zone or hippocampus of the transgenic compared to wild type mouse brains. By contrast, no such decreases were evident with the mdr1a isoform of P-glycoprotein and Bcrp, transporters predominantly found in brain endothelium. Such transporter expression changes could not be accounted for by alterations in blood vessel density or neuronal to glial cell ratios as analyzed both from immunocytochemical staining and from RT-PCR. These observations support previous in vitro data showing that manipulations to GSK-3beta activity that alter signaling via beta-catenin can influence the expression of efflux transporters. Implications from this are that drug distribution into cells within the brain of these transgenic mice could be enhanced, hence warranting further investigation.


Assuntos
Encéfalo/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , beta Catenina/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Western Blotting , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Contagem de Células , Endotélio Vascular/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Neuroglia/metabolismo , Neurônios/metabolismo , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
16.
J Neurochem ; 106(4): 1855-65, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18624906

RESUMO

This study investigates involvement of beta-catenin signalling in regulation of p-glycoprotein (p-gp) expression in endothelial cells derived from brain vasculature. Pharmacological interventions that enhance or that block beta-catenin signalling were applied to primary rat brain endothelial cells and to immortalized human brain endothelial cells, hCMEC/D3, nuclear translocation of beta-catenin being determined by immunocytochemistry and by western blot analysis to confirm effectiveness of the manipulations. Using the specific glycogen synthase kinase-3 (GSK-3) inhibitor 6-bromoindirubin-3'-oxime enhanced beta-catenin and increased p-gp expression including activating the MDR1 promoter. These increases were accompanied by increases in p-gp-mediated efflux capability as observed from alterations in intracellular fluorescent calcein accumulation detected by flow cytometry. Similar increases in p-gp expression were noted with other GSK-3 inhibitors, i.e. 1-azakenpaullone or LiCl. Application of Wnt agonist [2-amino-4-(3,4-(methylenedioxy) benzylamino)-6-(3-methoxyphenyl)pyrimidine] also enhanced beta-catenin and increased transcript and protein levels of p-gp. By contrast, down-regulating the pathway using Dickkopf-1 or quercetin decreased p-gp expression. Similar changes were observed with multidrug resistance protein 4 and breast cancer resistance protein, both known to be present at the blood-brain barrier. These results suggest that regulation of p-gp and other multidrug efflux transporters in brain vasculature can be influenced by beta-catenin signalling.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Encéfalo/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/fisiologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Transdução de Sinais/fisiologia , beta Catenina/fisiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/fisiologia , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Linhagem Celular Transformada , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Células Endoteliais/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Indóis/farmacologia , Masculino , Oximas/farmacologia , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , beta Catenina/genética
17.
Pflugers Arch ; 456(5): 801-12, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18214525

RESUMO

Transport activities involved in intracellular pH (pH(i)) recovery after acid or alkali challenge were investigated in cultured rat brain microvascular endothelial cells by monitoring pH(i) using a pH-sensitive dye. Following relatively small acid loads with pH(i) approximately 6.5, HCO(-)(3) influx accounted for most of the acid extrusion from the cell with both Cl(-)-independent and Cl(-)-dependent, Na(+)-dependent transporters involved. The Cl(-)-independent component has the same properties as the NBC-like transporter previously shown to account for most of the acid extrusion near the resting pH(i). Following large acid loads with pH(i) < 6.5, most of the acid extrusion was mediated by Na(+)/H(+) exchange, the rate of which was steeply dependent on pH(i). Concanamycin A, an inhibitor of V-type ATPase, had no effect on the rates of acid extrusion. Following an alkali challenge, the major component of the acid loading leading to recovery of pH(i) occurred by Cl(-)/HCO(-)(3) exchange. This exchange had the same properties as the AE-like transporter previously identified as a major acid loader near resting pH(i). These acid-loading and acid-extruding transport mechanisms together with the Na(+), K(+), ATPase may be sufficient to account not only for pH(i) regulation in brain endothelial cells but also for the net secretion of HCO(-)(3) across the blood-brain barrier.


Assuntos
Ácidos/metabolismo , Álcalis/metabolismo , Transporte Biológico/fisiologia , Encéfalo/irrigação sanguínea , Células Endoteliais/metabolismo , Concentração de Íons de Hidrogênio , Microcirculação , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/metabolismo , Trifosfato de Adenosina/metabolismo , Amilorida/análogos & derivados , Amilorida/metabolismo , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Carbonatos/metabolismo , Células Cultivadas , Cloretos/metabolismo , Desoxiglucose/metabolismo , Células Endoteliais/citologia , Fármacos Neuroprotetores/metabolismo , Ratos , Ratos Wistar , Sódio/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo
18.
Pflugers Arch ; 456(2): 379-91, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18026984

RESUMO

The endothelial cells of the brain microvasculature, which constitute the blood-brain barrier, secrete K+ into brain interstitial fluid. K+ channels are predicted to have a central role to play in this process. The aim of the following study was to characterise K+ channels in primary cultures of endothelial cells isolated from rat brain microvessels by whole-cell patch clamp and real-time polymerase chain reaction. In the 4 h after plating, the rat brain endothelial cells expressed predominantly a depolarisation-activated delayed-rectifying outward K+ conductance and a time-independent inwardly rectifying K+ conductance prominent at hyperpolarising potentials. The outward current was inhibited by 1 mM 4-aminopyridine (4AP), 10 nM margatoxin and 100 nM dendrotoxin-K, indicating the involvement of Kv1 channels. The half maximal activation voltage and time constants of activation and inactivation of the 4AP-sensitive current were similar to Kv1.3. The inwardly rectifying conductance was inhibited by Ba2+ in a dose- and voltage-dependent fashion; the kinetics of which resembled Kir2 channels. Quantification of messenger ribonucleic acid transcripts revealed Kv1.3>1.2=1.4=1.5=1.6 and Kir2.1=2>2.3. In current-clamp experiments, both 4AP and Ba2+ depolarised the membrane potential. In conclusion, rat brain endothelial cells express Kv1 and Kir2 K+ channels, both of which participate in setting membrane potential and could mediate K+ secretion into the brain interstitial fluid.


Assuntos
Encéfalo/irrigação sanguínea , Endotélio Vascular/metabolismo , Canal de Potássio Kv1.3/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , 4-Aminopiridina/farmacologia , Animais , Bário/farmacologia , Barreira Hematoencefálica/metabolismo , Plexo Corióideo/metabolismo , Endotélio Vascular/citologia , Masculino , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Venenos de Escorpião/farmacologia
19.
Brain Res ; 1159: 67-76, 2007 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-17583679

RESUMO

This study explores the effects of neural precursor cells (NPCs) on barrier characteristics in brain vasculature. Primary rat brain endothelial cells were exposed to conditioned medium from NPCs isolated from day 14 embryonic rat brains and maintained as free-floating undifferentiated neurospheres. Such exposure increased brain endothelial transcript levels of the mdr1a but not mdr1b gene encoding P-glycoprotein (Pgp) and reduced proliferation but did not alter transendothelial resistance (TER). These effects were compared to those seen following co-culture with differentiating NPCs or with primary astrocytes. NPCs, if grown adherent, differentiate into glial and neuronal cells as assessed by immunocytochemical and mRNA analysis. Brain endothelial cells when co-cultured with these cells also showed reduced proliferation and enhanced mdr1a expression, but in addition increased TER. Similar increases were observed in co-culture with astrocytes. These results suggest that undifferentiated NPCs produce factors that influence Pgp expression whereas their progeny also affect tight junction integrity.


Assuntos
Encéfalo/citologia , Diferenciação Celular/fisiologia , Células Endoteliais/fisiologia , Neurônios/fisiologia , Células-Tronco/fisiologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Astrócitos/fisiologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura/métodos , Meios de Cultivo Condicionados/farmacologia , Embrião de Mamíferos , Regulação da Expressão Gênica/fisiologia , Masculino , Ratos , Ratos Wistar , Trítio/farmacocinética , Vincristina/farmacocinética
20.
J Physiol ; 576(Pt 3): 769-85, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16916905

RESUMO

Fluid secretion across the blood-brain barrier, critical for maintaining the correct fluid balance in the brain, entails net secretion of HCO(3)(-), which is brought about by the combined activities of ion transporters situated in brain microvessels. These same transporters will concomitantly influence intracellular pH (pH(i)). To analyse the transporters that may be involved in the maintenance of pH(i) and hence secretion of HCO(3)(-), we have loaded primary cultured endothelial cells derived from rat brain microvessels with the pH indicator BCECF and suspended them in standard NaCl solutions buffered with Hepes or Hepes plus 5% CO(2)/HCO(3)(-). pH(i) in the standard solutions showed a slow acidification over at least 30 min, the rate being less in the presence of HCO(3)(-) than in its absence. However, after accounting for the difference in buffering, the net rates of acid loading with and without HCO(3)(-) were similar. In the nominal absence of HCO(3)(-) the rate of acid loading was increased equally by removal of external Na(+) or by inhibition of Na(+)/H(+) exchange by ethylisopropylamiloride (EIPA). By contrast, in the presence of HCO(3)(-) the increase in the rate of acid loading when Na(+) was removed was much larger and the rate was then also significantly greater than the rate observed in the absence of both Na(+) and HCO(3)(-). Removal of Cl(-) in the presence of HCO(3)(-) produced an alkalinization followed by a resumption of the slow acid gain. Removal of Na(+) following removal of Cl(-) increased the rate of acid gain. In the presence of HCO(3)(-) and initial presence of Na(+) and Cl(-), DIDS inhibited the changes in pH(i) produced by removal of either Na(+) or Cl(-). These are the expected results if these cells possess an AE-like Cl(-)/HCO(3)(-) exchanger, a 'channel-like' permeability allowing slow influx of acid (or efflux of HCO(3)(-)), a NBC-like Cl(-)-independent Na(+)-HCO(3)(-) cotransporter, and a NHE-like Na(+)/H(+) exchanger. The in vitro rates of HCO(3)(-) loading via the Na(+)-HCO(3)(-) cotransporter could, if the transporter is located on the apical, blood-facing side of the cells, account for the net secretion of HCO(3)(-) into the brain.


Assuntos
Antiportadores de Cloreto-Bicarbonato/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Simportadores de Cloreto de Sódio/fisiologia , Simportadores de Sódio-Bicarbonato/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Amilorida/análogos & derivados , Amilorida/farmacologia , Cloreto de Amônio/farmacologia , Animais , Barreira Hematoencefálica/fisiologia , Células Cultivadas , Endotélio Vascular/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Matemática , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Wistar , ATPase Trocadora de Sódio-Potássio/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...