Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Biomembr ; 1862(11): 183430, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32750317

RESUMEN

The choroid plexus (CP) is located in the ventricular system of the brain (one in each ventricle), and the CP epithelial cells form an important barrier between the blood and the cerebrospinal fluid (CSF). Their main function comprises CSF secretion, maintenance of brain homeostasis, signalling, and forming a neuroprotective barrier against harmful external and internal compounds. The CPs mature early and demonstrate expressional changes of barrier-specific genes and proteins related to location and developmental stage of the CP. Important proteins for the barrier function include tight junction proteins, numerous transporters and enzymes. Natural senescence leads to structural changes in the CP cells and reduced or loss of function, while further loss of CP function and changes in immune status may be relevant in neurodegenerative diseases such as Alzheimer's disease and Multiple Sclerosis. Neuroprotective genes expressed at CPs may be unexplored targets for new therapies for neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer , Barrera Hematoencefálica , Plexo Coroideo , Células Epiteliales , Regulación de la Expresión Génica/inmunología , Esclerosis Múltiple , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Plexo Coroideo/inmunología , Plexo Coroideo/patología , Células Epiteliales/inmunología , Células Epiteliales/patología , Humanos , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología
2.
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
4.
Front Neurosci ; 8: 359, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25426016

RESUMEN

Clinical data continue to reveal that the incidence of perinatal stroke is high, similar to that in the elderly. Perinatal stroke leads to significant morbidity and severe long-term neurological and cognitive deficits, including cerebral palsy. Experimental models of cerebral ischemia in neonatal rodents have shown that the pathophysiology of perinatal brain damage is multifactorial. Cerebral vasculature undergoes substantial structural and functional changes during early postnatal brain development. Thus, the state of the vasculature could affect susceptibility of the neonatal brain to cerebral ischemia. In this review, we discuss some of the most recent findings regarding the neurovascular responses of the immature brain to focal arterial stroke in relation to neuroinflammation. We also discuss a possible role of the neonatal blood-CSF barrier in modulating inflammation and the long-term effects of early neurovascular integrity after neonatal stroke on angiogenesis and neurogenesis.

5.
Fluids Barriers CNS ; 10(1): 25, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23915922

RESUMEN

BACKGROUND: The choroid plexuses are the interface between the blood and the cerebrospinal fluid (CSF) contained within the ventricular spaces of the central nervous system. The tight junctions linking adjacent cells of the choroidal epithelium create a physical barrier to paracellular movement of molecules. Multispecific efflux transporters as well as drug-metabolizing and antioxidant enzymes functioning in these cells contribute to a metabolic barrier. These barrier properties reflect a neuroprotective function of the choroid plexus. The choroid plexuses develop early during embryogenesis and provide pivotal control of the internal environment throughout development when the brain is especially vulnerable to toxic insults. Perinatal injuries like hypoxia and trauma, and exposure to drugs or toxic xenobiotics can have serious consequences on neurogenesis and long-term development. The present study describes the developmental expression pattern of genes involved in the neuroprotective functions of the blood-CSF barrier. METHODS: The transcriptome of rat lateral ventricular choroid plexuses isolated from fifteen-day-old embryos, nineteen-day old fetuses, two-day old pups, and adults was analyzed by a combination of Affymetrix microarrays, Illumina RNA-Sequencing, and quantitative RT-PCR. RESULTS: Genes coding for proteins involved in junction formation are expressed early during development. Overall perinatal expression levels of genes involved in drug metabolism and antioxidant mechanisms are similar to, or higher than levels measured in adults. A similar developmental pattern was observed for multispecific efflux transporter genes of the Abc and Slc superfamilies. Expression of all these genes was more variable in choroid plexus from fifteen-day-old embryos. A large panel of transcription factors involved in the xenobiotic- or cell stress-mediated induction of detoxifying enzymes and transporters is also expressed throughout development. CONCLUSIONS: This transcriptomic analysis suggests relatively well-established neuroprotective mechanisms at the blood-CSF barrier throughout development of the rat. The expression of many transcription factors early in development raises the possibility of additional protection for the vulnerable developing brain, should the fetus or newborn be exposed to drugs or other xenobiotics.

6.
PLoS One ; 8(7): e65629, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23843944

RESUMEN

We provide comprehensive identification of embryonic (E15) and adult rat lateral ventricular choroid plexus transcriptome, with focus on junction-associated proteins, ionic influx transporters and channels. Additionally, these data are related to new structural and previously published permeability studies. Results reveal that most genes associated with intercellular junctions are expressed at similar levels at both ages. In total, 32 molecules known to be associated with brain barrier interfaces were identified. Nine claudins showed unaltered expression, while two claudins (6 and 8) were expressed at higher levels in the embryo. Expression levels for most cytoplasmic/regulatory adaptors (10 of 12) were similar at the two ages. A few junctional genes displayed lower expression in embryos, including 5 claudins, occludin and one junctional adhesion molecule. Three gap junction genes were enriched in the embryo. The functional effectiveness of these junctions was assessed using blood-delivered water-soluble tracers at both the light and electron microscopic level: embryo and adult junctions halted movement of both 286Da and 3kDa molecules into the cerebrospinal fluid (CSF). The molecular identities of many ion channel and transporter genes previously reported as important for CSF formation and secretion in the adult were demonstrated in the embryonic choroid plexus (and validated with immunohistochemistry of protein products), but with some major age-related differences in expression. In addition, a large number of previously unidentified ion channel and transporter genes were identified for the first time in plexus epithelium. These results, in addition to data obtained from electron microscopical and physiological permeability experiments in immature brains, indicate that exchange between blood and CSF is mainly transcellular, as well-formed tight junctions restrict movement of small water-soluble molecules from early in development. These data strongly indicate the brain develops within a well-protected internal environment and the exchange between the blood, brain and CSF is transcellular and not through incomplete barriers.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Proteínas Portadoras/genética , Plexo Coroideo/metabolismo , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Transcriptoma , Animales , Barrera Hematoencefálica/citología , Proteínas Portadoras/metabolismo , Plexo Coroideo/citología , Claudinas/genética , Claudinas/metabolismo , Embrión de Mamíferos , Células Epiteliales/citología , Femenino , Perfilación de la Expresión Génica , Inmunohistoquímica , Uniones Intercelulares/genética , Uniones Intercelulares/metabolismo , Transporte Iónico , Microscopía Electrónica , Ocludina/genética , Ocludina/metabolismo , Permeabilidad , Embarazo , Ratas , Ratas Sprague-Dawley
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.
Am J Surg Pathol ; 36(6): 916-28, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22588068

RESUMEN

Neuroepithelial papillary tumor of the pineal region (PTPR) has been defined as a distinct entity that is increasingly being recognized, with 96 cases now reported. This tumor shares morphologic features with both ependymomas and choroid plexus tumors. PTPR is characterized by an epithelial-like growth pattern in which the vessels are covered by layers of tumor cells forming perivascular pseudorosettes. These tumors exhibit various combinations of papillary and solid architecture, making the differential diagnosis of PTPR difficult to establish. We report the detailed description of the histopathologic features of a large series of PTPRs from 20 different centers and distinguish 2 subgroups of tumors with either a striking papillary growth pattern or a papillary and solid growth pattern. We highlight the findings that PTPRs have unusual vessels with multiple lumina and frequently show detachment of the border of the tumoral cells from the vascular wall. The 2 PTPR subgroups present similar clinical characteristics and immunophenotypes. We confirmed and extended the results of previous ultrastructural studies on the presence of intercellular junctions at the apical part of tumoral cells. The expression of the tight junction proteins claudin-1, claudin-2, and claudin-3 was investigated by immunohistochemistry. Claudin-1 and claudin-3, but not claudin-2, were expressed in PTPRs and in the fetal subcommissural organ, potentially the origin of this tumor. In contrast, all 3 claudins were expressed in choroid plexus papillomas. Claudin expression may help in the diagnosis of PTPRs and can be used in combination with other markers, such as CK18, NCAM, E-cadherin, MAP-2, and Kir 7.1.


Asunto(s)
Carcinoma Papilar/patología , Claudinas/metabolismo , Pinealoma/patología , Uniones Estrechas/metabolismo , Adolescente , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Carcinoma Papilar/diagnóstico por imagen , Carcinoma Papilar/metabolismo , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pinealoma/metabolismo , Pinealoma/cirugía , Uniones Estrechas/ultraestructura , Ultrasonografía , Adulto Joven
9.
Free Radic Biol Med ; 48(12): 1588-600, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20226853

RESUMEN

Recent observations link myeloperoxidase (MPO) activation to neurodegeneration. In multiple sclerosis MPO is present in areas of active demyelination where the potent oxidant hypochlorous acid (HOCl), formed by MPO from H(2)O(2) and chloride ions, could oxidatively damage myelin-associated lipids. The purpose of this study was (i) to characterize reaction products of sphingomyelin (SM) formed in response to modification by HOCl, (ii) to define the impact of exogenously added SM and HOCl-modified SM (HOCl-SM) on viability parameters of a neuronal cell line (PC12), and (iii) to study alterations in the PC12 cell proteome in response to SM and HOCl-SM. MALDI-TOF-MS analyses revealed that HOCl, added as reagent or generated enzymatically, transforms SM into chlorinated species. On the cellular level HOCl-SM but not SM induced the formation of reactive oxygen species. HOCl-SM induced severely impaired cell viability, dissipation of the mitochondrial membrane potential, and activation of caspase-3 and DNA damage. Proteome analyses identified differential expression of specific subsets of proteins in response to SM and HOCl-SM. Our results demonstrate that HOCl modification of SM results in the generation of chlorinated lipid species with potent neurotoxic properties. Given the emerging connections between the MPO-H(2)O(2)-chloride axis and neurodegeneration, this chlorinating pathway might be implicated in neuropathogenesis.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácido Hipocloroso/farmacología , Neuronas/metabolismo , Oxidantes/farmacología , Estrés Oxidativo/fisiología , Esfingomielinas/metabolismo , Animales , Apoptosis/fisiología , Western Blotting , Química Encefálica , Línea Celular , Dopamina/metabolismo , Electroforesis en Gel Bidimensional , Análisis de Fourier , Halogenación , Procesamiento de Imagen Asistido por Computador , Técnicas In Vitro , Lípidos/química , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Proteoma/química , Proteoma/efectos de los fármacos , Proteoma/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Esfingomielinas/química
10.
J Neurochem ; 108(3): 707-18, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19046407

RESUMEN

Alpha-tocopherol (alphaTocH), a member of the vitamin E family, is essential for normal neurological function. Despite the importance of alphaTocH transport into the CNS, transfer mechanisms across the blood-brain barrier (BBB) are not entirely clear. We here investigate whether afamin, a known alphaTocH-binding protein, contributes to alphaTocH transport across an in vitro model of the BBB consisting of primary porcine brain capillary endothelial cells (BCEC) and basolaterally cultured astrocytoma cells. Exogenously added afamin had no adverse effects on BCEC viability or barrier function and was transported across BCEC Transwell cultures. Furthermore, alphaTocH transport across polarized BCEC cultures to astrocytoma cells is facilitated by afamin, though to a lesser extent than by high-density lipoprotein-mediated transport, an essential and in vivo operating alphaTocH import pathway at the cerebrovasculature. We also demonstrate that porcine BCEC endogenously synthesize afamin. In line with these in vitro findings, afamin was detected by immunohistochemistry in porcine, human postmortem, and mouse brain, where prominent staining was observed almost exclusively in the cerebrovasculature. The demonstration of afamin mRNA expression in isolated brain capillaries suggests that afamin might be a new family member of binding/transport proteins contributing to alphaTocH homeostasis at the BBB in vivo.


Asunto(s)
Barrera Hematoencefálica/fisiología , Proteínas Portadoras/biosíntesis , Circulación Cerebrovascular/fisiología , Células Endoteliales/metabolismo , Glicoproteínas/biosíntesis , Albúmina Sérica/biosíntesis , alfa-Tocoferol/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitoma/metabolismo , Transporte Biológico Activo , Western Blotting , Células CHO , Capilares/metabolismo , Técnicas de Cocultivo , Cricetinae , Cricetulus , Electroforesis en Gel de Poliacrilamida , Técnica del Anticuerpo Fluorescente , Humanos , Lipoproteínas HDL/biosíntesis , Lipoproteínas HDL/aislamiento & purificación , Ratones , Ratones Endogámicos C57BL , ARN/biosíntesis , ARN/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Albúmina Sérica Humana , Porcinos , Sales de Tetrazolio , Tiazoles
11.
J Control Release ; 117(3): 301-11, 2007 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-17239472

RESUMEN

Drug delivery to the brain is severely restricted by formation of tight junctions between adjacent brain capillary endothelial cells (BCEC). In the present study we have evaluated the effects of protamine-oligonucleotide nanoparticles (proticles) on the functional properties of primary porcine BCEC and characterized uptake and transcytosis of proticles by these cells. Proticles had no adverse effects on BCEC properties relevant to blood-brain barrier (BBB) function. Transcytosis of (125)I-labeled proticles across polarized BCEC cultures occurred in a time- and concentration-dependent manner. As apolipoproteins were suggested to enhance cellular proticle uptake, proticle coating was performed with apoA-I, the major apolipoprotein component of high density lipoproteins. Adsorption of apoA-I on the surface of proticles resulted in significantly improved uptake and transcytosis properties as compared to uncoated proticles. ApoA-I coating enhanced proticle delivery to astrocytes in an in vitro model of the BBB almost twofold. Blocking of scavenger receptor class B, type I (the prime receptor for high density lipoprotein/apoA-I that is expressed on BCEC) reduced transcytosis of apoA-I-coated proticles to levels observed for uncoated proticles. Our data indicate that apoA-I-coating of proticles could be a feasible targeting technology to improve delivery across the BBB.


Asunto(s)
Apolipoproteína A-I/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Nanopartículas , Oligonucleótidos/farmacología , Protaminas/farmacología , Animales , Astrocitos/metabolismo , Western Blotting , Encéfalo/citología , Química Encefálica/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Humanos , Lipoproteínas HDL/química , Lipoproteínas HDL/aislamiento & purificación , Tamaño de la Partícula , Espectrometría de Masa por Ionización de Electrospray , Porcinos , Sales de Tetrazolio , Tiazoles
12.
Int J Biochem Cell Biol ; 38(8): 1314-29, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16530456

RESUMEN

The blood-brain barrier contributes to maintain brain cholesterol metabolism and protects this uniquely balanced system from exchange with plasma lipoprotein cholesterol. Brain capillary endothelial cells, representing a physiological barrier to the central nervous system, express apolipoprotein A-I (apoA-I, the major high-density lipoprotein (HDL)-associated apolipoprotein), ATP-binding cassette transporter A1 (ABCA1), and scavenger receptor, class B, type I (SR-BI), proteins that promote cellular cholesterol mobilization. Liver X receptors (LXRs) and peroxisome-proliferator activated receptors (PPARs) are regulators of cholesterol transport, and activation of LXRs and PPARs has potential therapeutic implications for lipid-related neurodegenerative diseases. To clarify the functional impact of LXR/PPAR activation, sterol transport along the: (i) ABCA1/apoA-I and (ii) SR-BI/HDL pathway was investigated in primary, polarized brain capillary endothelial cells, an in vitro model of the blood-brain barrier. Activation of LXR (24(S)OH-cholesterol, TO901317), PPARalpha (bezafibrate, fenofibrate), and PPARgamma (troglitazone, pioglitazone) modulated expression of apoA-I, ABCA1, and SR-BI on mRNA and/or protein levels without compromising transendothelial electrical resistance or tight junction protein expression. LXR-agonists and troglitazone enhanced basolateral-to-apical cholesterol mobilization in the absence of exogenous sterol acceptors. Along with the induction of cell surface-located ABCA1, several agonists enhanced cholesterol mobilization in the presence of exogenous apoA-I, while efflux of 24(S)OH-cholesterol (the major brain cholesterol metabolite) in the presence of exogenous HDL remained unaffected. Summarizing, in cerebrovascular endothelial cells apoA-I, ABCA1, and SR-BI represent drug targets for LXR and PPAR-agonists to interfere with cholesterol homeostasis at the periphery of the central nervous system.


Asunto(s)
Polaridad Celular/fisiología , Proteínas de Unión al ADN/agonistas , Endotelio Vascular/metabolismo , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Citoplasmáticos y Nucleares/agonistas , Esteroles/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Células Cultivadas , Ácido Clofíbrico/síntesis química , Ácido Clofíbrico/farmacología , Proteínas de Unión al ADN/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Immunoblotting , Lipoproteínas HDL/metabolismo , Lipoproteínas HDL3 , Receptores X del Hígado , Microscopía Fluorescente , Modelos Biológicos , Receptores Nucleares Huérfanos , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Pioglitazona , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Depuradores de Clase B/genética , Receptores Depuradores de Clase B/metabolismo , Transducción de Señal/efectos de los fármacos , Esteroles/química , Porcinos , Tiazolidinedionas/síntesis química , Tiazolidinedionas/farmacología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética
13.
J Neurochem ; 94(1): 109-19, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15953354

RESUMEN

Normal neurological function depends on a constant supply of polyunsaturated fatty acids to the brain. A considerable proportion of essential fatty acids originates from lipoprotein-associated lipids that undergo uptake and/or catabolism at the blood-brain barrier (BBB). This study aimed at identifying expression and regulation of endothelial lipase (EL) in brain capillary endothelial cells (BCEC), major constituents of the BBB. Our results revealed that BCEC are capable of EL synthesis and secretion. Overexpression of EL resulted in enhanced hydrolysis of extracellular high-density lipoprotein (HDL)-associated sn-2-labeled [(14)C]20 : 4 phosphatidylcholine. [(14)C]20 : 4 was recovered in cellular lipids, indicating re-uptake and intracellular re-esterification. To investigate local regulation of EL in the cerebrovasculature, BCEC were cultured in the presence of peroxisome-proliferator activated receptor (PPAR)- and liver X receptor (LXR)-agonists, known to regulate HDL levels. These experiments revealed that 24(S)OH-cholesterol (a LXR agonist), bezafibrate (a PPARalpha agonist), or pioglitazone (a PPARgamma agonist) resulted in down-regulation of EL mRNA and protein levels. Our findings implicate that EL could generate fatty acids at the BBB for transport to deeper regions of the brain as building blocks for membrane phospholipids. In addition PPAR and LXR agonists appear to contribute to HDL homeostasis at the BBB by regulating EL expression.


Asunto(s)
Barrera Hematoencefálica/enzimología , Encéfalo/enzimología , Endotelio Vascular/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Lipasa/biosíntesis , Animales , Células Cultivadas , Humanos , Lipasa/genética , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...