Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neurol Sci ; 450: 120663, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37182424

RESUMEN

Neurological infections, such as Cerebral malaria (CM) and meningitis are associated with high mortality and in survivors, particularly young children, persistent neurologic deficits often remain. As brain inflammation plays a role in the development of these neurological sequelae, multiplex assays were used to assess a select set of immune mediators in both plasma and cerebrospinal fluid (CSF) from Zambian children with neurological infections. Both CM and meningitis patients showed high levels of markers for vascular inflammation, such as soluble ICAM-1 and angiopoietins. Although high levels of angiopoietin 1 and angiopoietin 2 were found in the meningitis group, their levels in the CSF were low and did not differ. As expected, there were high levels of cytokines and notably a significantly elevated IL-6 level in the CSF of the meningitis group. Interestingly, although elevated levels BDNF were found, BDNF levels were significantly higher in plasma of the meningitis group but similar in the CSF. The striking differences in plasma BDNF and IL-6 levels in the CSF point to markedly different neuro-pathological processes. Therefore, further investigations in the role of both IL-6 and BDNF in the neurological outcomes are needed.


Asunto(s)
Malaria Cerebral , Meningitis , Niño , Preescolar , Humanos , Factor Neurotrófico Derivado del Encéfalo , Citocinas/líquido cefalorraquídeo , Interleucina-6/líquido cefalorraquídeo , Malaria Cerebral/líquido cefalorraquídeo , Meningitis/líquido cefalorraquídeo
2.
Int J Mol Sci ; 24(8)2023 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-37108071

RESUMEN

Postmortem neuropathology shows clear regional differences in many brain diseases. For example, brains from cerebral malaria (CM) patients show more hemorrhagic punctae in the brain's white matter (WM) than grey matter (GM). The underlying reason for these differential pathologies is unknown. Here, we assessed the effect of the vascular microenvironment on brain endothelial phenotype, focusing endothelial protein C receptor (EPCR). We demonstrate that the basal level of EPCR expression in cerebral microvessels is heterogeneous in the WM compared to the GM. We used in vitro brain endothelial cell cultures and showed that the upregulation of EPCR expression was associated with exposure to oligodendrocyte conditioned media (OCM) compared to astrocyte conditioned media (ACM). Our findings shed light on the origin of the heterogeneity of molecular phenotypes at the microvascular level and might help better understand the variation in pathology seen in CM and other neuropathologies associated with vasculature in various brain regions.


Asunto(s)
Astrocitos , Receptor de Proteína C Endotelial , Malaria Cerebral , Humanos , Astrocitos/metabolismo , Encéfalo/metabolismo , Medios de Cultivo Condicionados/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Endotelio/metabolismo , Oligodendroglía/metabolismo
3.
Malar J ; 19(1): 266, 2020 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-32703204

RESUMEN

Cerebral malaria (CM), results from Plasmodium falciparum infection, and has a high mortality rate. CM survivors can retain life-long post CM sequelae, including seizures and neurocognitive deficits profoundly affecting their quality of life. As the Plasmodium parasite does not enter the brain, but resides inside erythrocytes and are confined to the lumen of the brain's vasculature, the neuropathogenesis leading to these neurologic sequelae is unclear and under-investigated. Interestingly, postmortem CM pathology differs in brain regions, such as the appearance of haemorragic punctae in white versus gray matter. Various host and parasite factors contribute to the risk of CM, including exposure at a young age, parasite- and host-related genetics, parasite sequestration and the extent of host inflammatory responses. Thus far, several proposed adjunctive treatments have not been successful in the treatment of CM but are highly needed. The region-specific CM neuro-pathogenesis leading to neurologic sequelae is intriguing, but not sufficiently addressed in research. More attention to this may lead to the development of effective adjunctive treatments to address CM neurologic sequelae.


Asunto(s)
Malaria Cerebral , Malaria Falciparum , Plasmodium falciparum/fisiología , Eritrocitos/parasitología , Humanos , Malaria Cerebral/complicaciones , Malaria Cerebral/fisiopatología , Malaria Falciparum/complicaciones , Malaria Falciparum/fisiopatología , Calidad de Vida
4.
Front Cell Neurosci ; 13: 405, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31616251

RESUMEN

The blood-brain barrier (BBB) helps maintain a tightly regulated microenvironment for optimal central nervous system (CNS) homeostasis and facilitates communications with the peripheral circulation. The brain endothelial cells, lining the brain's vasculature, maintain close interactions with surrounding brain cells, e.g., astrocytes, pericytes and perivascular macrophages. This function facilitates critical intercellular crosstalk, giving rise to the concept of the neurovascular unit (NVU). The steady and appropriate communication between all components of the NVU is essential for normal CNS homeostasis and function, and dysregulation of one of its constituents can result in disease. Among the different brain regions, and along the vascular tree, the cellular composition of the NVU varies. Therefore, differential cues from the immediate vascular environment can affect BBB phenotype. To support the fluctuating metabolic and functional needs of the underlying neuropil, a specialized vascular heterogeneity is required. This is achieved by variances in barrier function, expression of transporters, receptors, and adhesion molecules. This mini-review will take you on a journey through evolving concepts surrounding the BBB, the NVU and beyond. Exploring classical experiments leading to new approaches will allow us to understand that the BBB is not merely a static separation between the brain and periphery but a closely regulated and interactive entity. We will discuss shifting paradigms, and ultimately aim to address the importance of BBB endothelial heterogeneity with regard to the function of the BBB within the NVU, and touch on its implications for different neuropathologies.

5.
PLoS One ; 14(3): e0213428, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30849122

RESUMEN

Clinical and model studies indicate that low nitric oxide (NO) bioavailability due in part to profound hypoargininemia contributes to cerebral malaria (CM) pathogenesis. Protection against CM pathogenesis may be achieved by altering the diet before infection with Plasmodium falciparum infection (nutraceutical) or by administering adjunctive therapy that decreases CM mortality (adjunctive therapy). This hypothesis was tested by administering citrulline or arginine in experimental CM (eCM). We report that citrulline injected as prophylaxis immediately post infection (PI) protected virtually all mice by ameliorating (i) hypoargininemia, (ii) urea cycle impairment, and (iii) disruption of blood brain barrier. Citrulline prophylaxis inhibited plasma arginase activity. Parasitemia was similar in citrulline- and vehicle control-groups, indicating that protection from pathogenesis was not due to decreased parasitemia. Both citrulline and arginine administered from day 1 PI in the drinking water significantly protected mice from eCM. These observations collectively indicate that increasing dietary citrulline or arginine decreases eCM mortality. Citrulline injected ip on day 4 PI with quinine-injected ip on day 6 PI partially protected mice from eCM; citrulline plus scavenging of superoxide with pegylated superoxide dismutase and pegylated catalase protected all recipients from eCM. These findings indicate that ameliorating hypoargininemia with citrulline plus superoxide scavenging decreases eCM mortality.


Asunto(s)
Citrulina/farmacología , Malaria Cerebral/metabolismo , Malaria Cerebral/prevención & control , Animales , Arginasa/sangre , Arginina/administración & dosificación , Arginina/sangre , Arginina/deficiencia , Barrera Hematoencefálica/efectos de los fármacos , Citrulina/administración & dosificación , Suplementos Dietéticos , Modelos Animales de Enfermedad , Depuradores de Radicales Libres/administración & dosificación , Humanos , Malaria Cerebral/etiología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Plasmodium berghei , Superóxidos/metabolismo , Urea/metabolismo
6.
Fluids Barriers CNS ; 15(1): 12, 2018 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-29688865

RESUMEN

The vertebrate blood-brain barrier (BBB) is composed of cerebral microvascular endothelial cells (CEC). The BBB acts as a semi-permeable cellular interface that tightly regulates bidirectional molecular transport between blood and the brain parenchyma in order to maintain cerebral homeostasis. The CEC phenotype is regulated by a variety of factors, including cells in its immediate environment and within functional neurovascular units. The cellular composition of the brain parenchyma surrounding the CEC varies between different brain regions; this difference is clearly visible in grey versus white matter. In this review, we discuss evidence for the existence of brain vascular heterogeneity, focusing on differences between the vessels of the grey and white matter. The region-specific differences in the vasculature of the brain are reflective of specific functions of those particular brain areas. This BBB-endothelial heterogeneity may have implications for the course of pathogenesis of cerebrovascular diseases and neurological disorders involving vascular activation and dysfunction. This heterogeneity should be taken into account when developing BBB-neuro-disease models representative of specific brain areas.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Sustancia Gris/irrigación sanguínea , Sustancia Gris/fisiopatología , Sustancia Blanca/irrigación sanguínea , Sustancia Blanca/fisiopatología , Animales , Humanos , Técnicas In Vitro , Modelos Biológicos , Acoplamiento Neurovascular/fisiología
7.
Cell Microbiol ; 17(12): 1883-99, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26119044

RESUMEN

Plasmodium falciparum-infected erythrocytes (IRBC) expressing the domain cassettes (DC) 8 and 13 of the cytoadherent ligand P. falciparum erythrocyte membrane protein 1 adhere to the endothelial protein C receptor (EPCR). By interfering with EPCR anti-coagulant and pro-endothelial barrier functions, IRBC adhesion could promote coagulation and vascular permeability that contribute to the pathogenesis of cerebral malaria. In this study, we examined the adhesion of DC8- and DC13-expressing parasite lines to endothelial cells from different microvasculature, and the consequences of EPCR engagement on endothelial cell function. We found that IRBC from IT4var19 (DC8) and IT4var07 (DC13) parasite lines adhered to human brain, lung and dermal endothelial cells under shear stress. However, the relative contribution of EPCR to parasite cytoadherence on different types of endothelial cell varied. We also observed divergent functional outcomes for DC8 cysteine-rich interdomain region (CIDR)α1.1 and DC13 CIDRα1.4 domains. IT4var07 CIDRα1.4 inhibited generation of activated protein C (APC) on lung and dermal endothelial cells and blocked the APC-EPCR binding interaction on brain endothelial cells. IT4var19 CIDRα1.1 inhibited thrombin-induced endothelial barrier dysfunction in lung endothelial cells, whereas IT4var07 CIDRα1.4 inhibited the protective effect of APC on thrombin-induced permeability. Overall, these findings reveal a much greater complexity of how CIDRα1-expressing parasites may modulate malaria pathogenesis through EPCR adhesion.


Asunto(s)
Antígenos CD/metabolismo , Adhesión Celular , Células Endoteliales/fisiología , Eritrocitos/parasitología , Interacciones Huésped-Patógeno , Plasmodium falciparum/fisiología , Receptores de Superficie Celular/metabolismo , Células Cultivadas , Receptor de Proteína C Endotelial , Humanos , Ligadura , Resultado del Tratamiento
8.
Proc Natl Acad Sci U S A ; 109(26): E1782-90, 2012 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-22619321

RESUMEN

Cerebral malaria (CM) is a deadly complication of Plasmodium falciparum infection, but specific interactions involved in cerebral homing of infected erythrocytes (IEs) are poorly understood. In this study, P. falciparum-IEs were characterized for binding to primary human brain microvascular endothelial cells (HBMECs). Before selection, CD36 or ICAM-1-binding parasites exhibited punctate binding to a subpopulation of HBMECs and binding was CD36 dependent. Panning of IEs on HBMECs led to a more dispersed binding phenotype and the selection of three var genes, including two that encode the tandem domain cassette 8 (DC8) and were non-CD36 binders. Multiple domains in the DC8 cassette bound to brain endothelium and the cysteine-rich interdomain region 1 inhibited binding of P. falciparum-IEs by 50%, highlighting a key role for the DC8 cassette in cerebral binding. It is mysterious how deadly binding variants are maintained in the parasite population. Clonal parasite lines expressing the two brain-adherent DC8-var genes did not bind to any of the known microvascular receptors, indicating unique receptors are involved in cerebral binding. They could also adhere to brain, lung, dermis, and heart endothelial cells, suggesting cerebral binding variants may have alternative sequestration sites. Furthermore, young African children with CM or nonsevere control cases had antibodies to HBMEC-selected parasites, indicating they had been exposed to related variants during childhood infections. This analysis shows that specific P. falciparum erythrocyte membrane protein 1 types are linked to cerebral binding and suggests a potential mechanism by which individuals may build up immunity to severe disease, in the absence of CM.


Asunto(s)
Encéfalo/irrigación sanguínea , Adhesión Celular , Endotelio Vascular/patología , Eritrocitos/parasitología , Genes Protozoarios , Malaria Cerebral/parasitología , Plasmodium falciparum/fisiología , Animales , Preescolar , Eritrocitos/patología , Humanos , Malaria Cerebral/patología , Plasmodium falciparum/genética
9.
Cell Microbiol ; 13(10): 1470-8, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21824246

RESUMEN

The vascular endothelium of the blood-brain barrier (BBB) is regarded as a part of the neurovascular unit (NVU). This emerging NVU concept emphasizes the need for homeostatic signalling among the neuronal, glial and vascular endothelial cellular compartments in maintaining normal brain function. Conversely, dysfunction in any component of the NVU affects another, thus contributing to disease. Brain endothelial activation and dysfunction is observed in various neurological diseases, such as (ischemic) stroke, seizure, brain inflammation and infectious diseases and likely contributes to or exacerbates neurological conditions. The role and impact of brain endothelial factors on astroglial and neuronal activation is unclear. Similarly, it is not clear which stages of BBB endothelial activation can be considered beneficial versus detrimental. Although the BBB plays an important role in context of encephalopathies caused by neurotropic microbes that must first penetrate into the brain, a crucial role of the BBB in contributing to neurological dysfunction may be seen in cerebral malaria (CM), where the Plasmodium parasite remains sequestered in the brain vasculature, does not enter the brain parenchyma, and yet causes coma and seizures. In this minireview some of the scenarios and factors that may play a role in BBB as a relay station to modulate astroneuronal functioning are discussed.


Asunto(s)
Astrocitos/fisiología , Barrera Hematoencefálica/fisiología , Células Endoteliales/fisiología , Interacciones Huésped-Patógeno , Neuronas/fisiología , Animales , Barrera Hematoencefálica/microbiología , Barrera Hematoencefálica/parasitología , Barrera Hematoencefálica/virología , Células Endoteliales/microbiología , Células Endoteliales/parasitología , Células Endoteliales/virología , Humanos
10.
Blood ; 114(19): 4243-52, 2009 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-19713460

RESUMEN

Cerebral malaria is a severe multifactorial condition associated with the interaction of high numbers of infected erythrocytes to human brain endothelium without invasion into the brain. The result is coma and seizures with death in more than 20% of cases. Because the brain endothelium is at the interface of these processes, we investigated the global gene responses of human brain endothelium after the interaction with Plasmodium falciparum-infected erythrocytes with either high- or low-binding phenotypes. The most significantly up-regulated transcripts were found in gene ontology groups comprising the immune response, apoptosis and antiapoptosis, inflammatory response, cell-cell signaling, and signal transduction and nuclear factor kappaB (NF-kappaB) activation cascade. The proinflammatory NF-kappaB pathway was central to the regulation of the P falciparum-modulated endothelium transcriptome. The proinflammatory molecules, for example, CCL20, CXCL1, CXCL2, IL-6, and IL-8, were increased more than 100-fold, suggesting an important role of blood-brain barrier (BBB) endothelium in the innate defense during P falciparum-infected erythrocyte (Pf-IRBC) sequestration. However, some of these diffusible molecules could have reversible effects on brain tissue and thus on neurologic function. The inflammatory pathways were validated by direct measurement of proteins in brain endothelial supernatants. This study delineates the strong inflammatory component of human brain endothelium contributing to cerebral malaria.


Asunto(s)
Malaria Cerebral/genética , Malaria Cerebral/fisiopatología , Malaria Falciparum/genética , Malaria Falciparum/fisiopatología , FN-kappa B/fisiología , Plasmodium falciparum/patogenicidad , Animales , Apoptosis , Barrera Hematoencefálica , Encéfalo/irrigación sanguínea , Encéfalo/parasitología , Células Cultivadas , Células Endoteliales/parasitología , Células Endoteliales/patología , Células Endoteliales/fisiología , Eritrocitos/parasitología , Eritrocitos/fisiología , Estudio de Asociación del Genoma Completo , Humanos , Inmunidad Innata , Inflamación/genética , Inflamación/parasitología , Inflamación/patología , Inflamación/fisiopatología , Malaria Cerebral/parasitología , Malaria Cerebral/patología , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal
11.
Stroke ; 40(2): 652-5, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18974377

RESUMEN

BACKGROUND AND PURPOSE: We recently reported that delayed lithium therapy can improve stroke recovery in rats by augmenting neurovascular remodeling. We tested the hypothesis that lithium can promote the expression of growth factors in brain endothelial cells and astrocytes. METHODS: Human brain microvascular endothelial cells and primary rat cortical astrocytes were exposed to lithium chloride in serum-free medium. We examined 2 representative growth factors: brain-derived neurotrophic factor and vascular endothelial growth factor (VEGF). Cell lysates were collected for Western blot analysis. Conditioned media was analyzed with enzyme-linked immunosorbent assay. SB-216763 and LY294002 were used to assess the roles of the glycogen synthase kinase-3beta (GSK-3beta) and PI3-K signaling in the lithium-induced responses. RESULTS: No consistent responses were observed for brain-derived neurotrophic factor. However, lithium (0.2 to 20 mmol/L) increased the phosphorylation of GSK-3beta and promoted VEGF secretion in a concentration-dependent manner in both endothelial and astrocyte cells. For endothelial cells, the potent GSK-3beta inhibitor SB-216763 upregulated VEGF, whereas inhibition of PI3-K with LY294002 suppressed lithium-induced responses in both phospho-GSK-3beta and VEGF. In contrast, neither inhibition of GSK-3beta nor inhibition of PI3-K had any detectable effects on VEGF levels in astrocytes. CONCLUSIONS: Lithium promotes VEGF expression through PI3-K/GSK-3beta-dependent and -independent pathways in brain endothelium and astrocytes, respectively. This growth factor signaling mechanism may contribute to lithium's reported ability to promote neurovascular remodeling after stroke.


Asunto(s)
Astrocitos/metabolismo , Química Encefálica/efectos de los fármacos , Células Endoteliales/metabolismo , Cloruro de Litio/farmacología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Astrocitos/efectos de los fármacos , Western Blotting , Encéfalo/citología , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Cromonas/farmacología , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/biosíntesis , Humanos , Indoles/farmacología , Maleimidas/farmacología , Morfolinas/farmacología , Regulación hacia Arriba/efectos de los fármacos
12.
Stroke ; 39(9): 2538-43, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18635843

RESUMEN

BACKGROUND AND PURPOSE: The concept of the neurovascular unit suggests that effects on brain vasculature must be considered if neuroprotection is to be achieved in stroke. We previously reported that 12/15-lipoxygenase (12/15-LOX) is upregulated in the peri-infarct area after middle cerebral artery occlusion in mice, and 12/15-LOX contributes to brain damage after ischemia-reperfusion. The current study was designed to investigate 12/15-LOX involvement in vascular injury in the ischemic brain. METHODS: In cell culture, a human brain microvascular endothelial cell line was subjected to either hypoxia or H(2)O(2)-induced oxidative stress with or without lipoxygenase inhibitors. For in vivo studies, mice were subjected to 90 minutes middle cerebral artery occlusion, and the effects of either 12/15-LOX gene knockout or treatment with lipoxygenase inhibitors were compared. Expression of 12/15-LOX and claudin-5 as well as extravasation of immunoglobulin G were detected by immunohistochemistry. Edema was measured as water content of brain hemispheres according to the wet-dry weight method. RESULTS: Brain endothelial cells were protected against hypoxia and H(2)O(2) by the lipoxygenase inhibitor baicalein. After focal ischemia, 12/15-LOX was increased in neurons and endothelial cells. The vascular tight junction protein claudin-5 underwent extensive degradation in the peri-infarct area, which was partially prevented by the lipoxygenase inhibitor baicalein. Leakage of immunoglobulin G into the brain parenchyma was significantly reduced in 12/15-LOX knockout mice as well as wild-type mice treated with baicalein. Likewise, brain edema was significantly ameliorated. CONCLUSIONS: 12/15-LOX may contribute to ischemic brain damage not just by causing neuronal cell death, but also by detrimental effects on the brain microvasculature. 12/15-LOX inhibitors may thus be effective as both neuroprotectants and vasculoprotectants.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/metabolismo , Edema Encefálico/enzimología , Infarto Encefálico/enzimología , Isquemia Encefálica/enzimología , Flavanonas/uso terapéutico , Ataque Isquémico Transitorio/enzimología , Animales , Araquidonato 12-Lipooxigenasa/efectos de los fármacos , Araquidonato 12-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/efectos de los fármacos , Araquidonato 15-Lipooxigenasa/genética , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Edema Encefálico/fisiopatología , Edema Encefálico/prevención & control , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/fisiopatología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/fisiopatología , Células Cultivadas , Claudina-5 , Citoprotección/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Peróxido de Hidrógeno/farmacología , Inmunoglobulina G/metabolismo , Ataque Isquémico Transitorio/tratamiento farmacológico , Ataque Isquémico Transitorio/fisiopatología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo
13.
Proc Natl Acad Sci U S A ; 105(21): 7582-7, 2008 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-18495934

RESUMEN

The neurovascular unit is an emerging concept that emphasizes homeostatic interactions between endothelium and cerebral parenchyma. Here, we show that cerebral endothelium are not just inert tubes for delivering blood, but they also secrete trophic factors that can be directly neuroprotective. Conditioned media from cerebral endothelial cells broadly protects neurons against oxygen-glucose deprivation, oxidative damage, endoplasmic reticulum stress, hypoxia, and amyloid neurotoxicity. This phenomenon is largely mediated by endothelial-produced brain-derived neurotrophic factor (BDNF) because filtering endothelial-conditioned media with TrkB-Fc eliminates the neuroprotective effect. Endothelial production of BDNF is sustained by beta-1 integrin and integrin-linked kinase (ILK) signaling. Noncytotoxic levels of oxidative stress disrupts ILK signaling and reduces endothelial levels of neuroprotective BDNF. These data suggest that cerebral endothelium provides a critical source of homeostatic support for neurons. Targeting these signals of matrix and trophic coupling between endothelium and neurons may provide new therapeutic opportunities for stroke and other CNS disorders.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Comunicación Celular , Endotelio Vascular/fisiología , Neuronas/fisiología , Animales , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Integrina beta1/metabolismo , Masculino , Ratones , Ratones Endogámicos , Neuronas/citología , Neuronas/metabolismo , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas/metabolismo
14.
J Infect Dis ; 195(7): 942-50, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17330783

RESUMEN

BACKGROUND: Central to the pathologic progression of human cerebral malaria (CM) is sequestration of Plasmodium falciparum-infected red blood cells (Pf-IRBCs) to the blood-brain barrier (BBB) endothelium. The molecular interactions between Pf-IRBCs and the BBB endothelium and their implications for barrier function are unclear. METHODS: The effects of Pf-IRBCs on the integrity of the BBB were assessed by electrical cell substrate sensing and by transendothelial electrical resistance measurements in an in vitro human BBB model. In addition, Pf-IRBCs were subfractionated and treated with trypsin, artemisinin, or brefeldin A. RESULTS: Pf-IRBCs, but not normal red blood cells, significantly decreased BBB resistance. Subfractionation showed that both membrane-associated and soluble Pf-IRBC factors mediate the decrease in BBB resistance. Trypsin treatment significantly reduced Pf-IRBC binding but not their ability to decrease electrical resistance. Likewise, P. falciparum isolates with increased binding to human brain microvascular endothelial cells did not alter the electrical resistance response. Soluble factors from Pf-IRBC culture supernatant decreased resistance by 50%-70% and precipitated with 40% ammonium sulfate saturation. Brefeldin-A partially blocked the ability of Pf-IRBCs to reduce resistance. CONCLUSION: The results suggest that, in CM, trypsin-resistant membrane components and soluble factors of Pf-IRBCs contribute to the impedance of BBB integrity in a multistep and multifactorial process.


Asunto(s)
Barrera Hematoencefálica/fisiología , Endotelio Vascular/parasitología , Eritrocitos/parasitología , Malaria Cerebral/parasitología , Plasmodium falciparum/fisiología , Animales , Encéfalo/parasitología , Impedancia Eléctrica , Endotelio Vascular/citología , Humanos
15.
Infect Immun ; 74(6): 3262-70, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16714553

RESUMEN

Sequestration of Plasmodium falciparum-infected erythrocytes (Pf-IRBC) in postcapillary brain endothelium is a hallmark of cerebral malaria (CM) pathogenesis. There is a correlation between adherent Pf-IRBC and increased expression of intercellular cell adhesion molecule 1 (ICAM-1), which is also a receptor for Pf-IRBC on human brain microvascular endothelial cells (HBMEC). The underlying mechanism for the increased ICAM-1 expression has not been clearly defined. Therefore, we investigated the mechanisms of ICAM-1 expression on isolated HBMEC after exposure to Pf-IRBC. Ultrastructural characterization of the model confirmed that there was attachment through both Pf-IRBC knobs and HBMEC microvillus protrusions. Pf-IRBC induced a dose- and time-dependent increase in ICAM-1 expression on HBMEC that was specific for human brain endothelium and was not observed with human umbilical vein endothelium. Involvement of both membrane-associated Pf-IRBC proteins and parasite-derived soluble factors with the increase in ICAM-1 expression was demonstrated by surface trypsinization and fractionation. Pf-IRBC exposure induced nuclear translocation of NF-kappaB in HBMEC, which was linked to ICAM-1 expression, as shown by use of specific inhibitors of the transcription factor NF-kappaB and immunocytochemistry. In addition, inhibition of reactive oxygen species decreased Pf-IRBC-induced ICAM-1 expression on HBMEC. Parasite-induced ICAM-1 expression explains the localization of this molecule on brain endothelium in postmortem CM brain samples. By increasing ICAM-1 expression, Pf-IRBC may increase their sequestration, thereby perpetuating CM.


Asunto(s)
Encéfalo/metabolismo , Endotelio Vascular/metabolismo , Eritrocitos/parasitología , Molécula 1 de Adhesión Intercelular/análisis , FN-kappa B/fisiología , Plasmodium falciparum/patogenicidad , Transporte Activo de Núcleo Celular , Animales , Artemisininas/farmacología , Barrera Hematoencefálica , Ensayo de Inmunoadsorción Enzimática , Humanos , Especies Reactivas de Oxígeno , Sesquiterpenos/farmacología
16.
BMC Cancer ; 6: 56, 2006 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-16524486

RESUMEN

BACKGROUND: Patients with metastatic tumors to the brain have a very poor prognosis. Increased metastatic potential has been associated with the fibrinolytic system. We investigated the role of the fibrinolytic enzyme plasmin in tumor cell migration across brain endothelial cells and growth of brain metastases in an experimental metastatic melanoma model. METHODS: Metastatic tumors to the brain were established by direct injection into the striatum or by intracarotid injection of B16F10 mouse melanoma cells in C57Bl mice. The role of plasminogen in the ability of human melanoma cells to cross a human blood-brain barrier model was studied on a transwell system. RESULTS: Wild type mice treated with the plasmin inhibitor epsilon-aminocaproic acid (EACA) and plg-/- mice developed smaller tumors and survived longer than untreated wild type mice. Tumors metastasized to the brain of wild type mice treated with EACA and plg-/- less efficiently than in untreated wild type mice. No difference was observed in the tumor growth in any of the three groups of mice. Human melanoma cells were able to cross the human blood-brain barrier model in a plasmin dependent manner. CONCLUSION: Plasmin facilitates the development of tumor metastasis to the brain. Inhibition of the fibrinolytic system could be considered as means to prevent tumor metastasis to the brain.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Neoplasias Encefálicas/secundario , Melanoma Experimental/secundario , Plasminógeno/fisiología , Neoplasias Cutáneas/patología , Ácido Aminocaproico/farmacología , Animales , Antifibrinolíticos/farmacología , Encéfalo/irrigación sanguínea , Encéfalo/citología , Neoplasias Encefálicas/fisiopatología , Arterias Carótidas , Línea Celular Tumoral , Movimiento Celular , Células Cultivadas , Endotelio Vascular/citología , Humanos , Inyecciones , Melanoma Experimental/patología , Melanoma Experimental/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica , Plasminógeno/genética , Neoplasias Cutáneas/fisiopatología
17.
Nat Med ; 12(2): 207-13, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16444266

RESUMEN

Natural killer (NK) cells and dendritic cells (DCs) are, respectively, central components of innate and adaptive immune responses. We describe here a third DC lineage, termed interferon-producing killer DCs (IKDCs), distinct from conventional DCs and plasmacytoid DCs and with the molecular expression profile of both NK cells and DCs. They produce substantial amounts of type I interferons (IFN) and interleukin (IL)-12 or IFN-gamma, depending on activation stimuli. Upon stimulation with CpG oligodeoxynucleotides, ligands for Toll-like receptor (TLR)-9, IKDCs kill typical NK target cells using NK-activating receptors. Their cytolytic capacity subsequently diminishes, associated with the loss of NKG2D receptor (also known as Klrk1) and its adaptors, Dap10 and Dap12. As cytotoxicity is lost, DC-like antigen-presenting activity is gained, associated with upregulation of surface major histocompatibility complex class II (MHC II) and costimulatory molecules, which formally distinguish them from classical NK cells. In vivo, splenic IKDCs preferentially show NK function and, upon systemic infection, migrate to lymph nodes, where they primarily show antigen-presenting cell activity. By virtue of their capacity to kill target cells, followed by antigen presentation, IKDCs provide a link between innate and adaptive immunity.


Asunto(s)
Células Dendríticas/inmunología , Interferones/biosíntesis , Células Asesinas Naturales/inmunología , Adaptación Fisiológica , Animales , Presentación de Antígeno , Línea Celular Tumoral , Citotoxicidad Inmunológica , Células Dendríticas/clasificación , Células Dendríticas/ultraestructura , Expresión Génica , Inmunidad Innata , Técnicas In Vitro , Células Asesinas Naturales/clasificación , Células Asesinas Naturales/ultraestructura , Listeria monocytogenes/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Oligodesoxirribonucleótidos/farmacología
18.
Brain ; 129(Pt 1): 212-23, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16230319

RESUMEN

Chemokines and chemokine receptors play a key role in the transmigration of leucocytes across the blood-brain barrier (BBB). CCR2 is the major receptor for CCL2, a potent monocyte and T cell chemoattractant. CCR2 and CCL2 have been consistently associated with a pathogenic role in experimental autoimmune encephalomyelitis, using knockout and transgenic mice, neutralizing antibodies, peptide antagonists and DNA vaccination. However, the significance of CCL2 and CCR2 in multiple sclerosis is enigmatic, because CCL2 levels are consistently decreased in the CSF of patients with this disease and other chronic neuroinflammatory conditions, despite abundant expression within lesional multiple sclerosis tissues. This study used an in vitro BBB model to test the hypothesis that CCL2 is removed from the extracellular fluid by CCR2-positive migrating cells as they cross the BBB, resulting in decreased CSF CCL2 levels. We showed that CCR2-positive T cells and monocytes migrated selectively across the in vitro BBB, and that CCL2 on the abluminal (tissue) side was consumed by migrating T cells and monocytes. Next, we used a new anti-CCR2 antibody to show that CCR2-positive mononuclear inflammatory cells could be readily detected in appropriate positive control tissues, but that CCR2+ cells were very infrequently found in multiple sclerosis lesions. We then showed that CCR2 receptor density on T cells and monocytes was specifically downregulated upon in vitro BBB transmigration in response to CCL2, but not irrelevant chemokines. These findings document a novel strategy for analysing chemokine receptor function in inflammatory CNS disease, and support the hypothesis that CCL2 is consumed by migrating inflammatory cells, which downregulate CCR2, as they cross the BBB.


Asunto(s)
Barrera Hematoencefálica , Quimiocina CCL2/metabolismo , Monocitos/metabolismo , Esclerosis Múltiple/inmunología , Receptores de Quimiocina/metabolismo , Linfocitos T/metabolismo , Anciano , Anciano de 80 o más Años , Movimiento Celular , Quimiocina CCL2/líquido cefalorraquídeo , Quimiotaxis de Leucocito , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática/métodos , Líquido Extracelular/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Microscopía Confocal , Persona de Mediana Edad , Esclerosis Múltiple/líquido cefalorraquídeo , Esclerosis Múltiple/metabolismo , Toxina del Pertussis/farmacología , Receptores CCR2
19.
Infect Immun ; 73(2): 1014-22, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15664945

RESUMEN

Neurological manifestations of Lyme disease in humans are attributed in part to penetration of the blood-brain barrier (BBB) and invasion of the central nervous system (CNS) by Borrelia burgdorferi. However, how the spirochetes cross the BBB remains an unresolved issue. We examined the traversal of B. burgdorferi across the human BBB and systemic endothelial cell barriers using in vitro model systems constructed of human brain microvascular endothelial cells (BMEC) and EA.hy 926, a human umbilical vein endothelial cell (HUVEC) line grown on Costar Transwell inserts. These studies showed that B. burgdorferi differentially crosses human BMEC and HUVEC and that the human BMEC form a barrier to traversal. During the transmigration by the spirochetes, it was found that the integrity of the endothelial cell monolayers was maintained, as assessed by transendothelial electrical resistance measurements at the end of the experimental period, and that B. burgdorferi appeared to bind human BMEC by their tips near or at cell borders, suggesting a paracellular route of transmigration. Importantly, traversal of B. burgdorferi across human BMEC induces the expression of plasminogen activators, plasminogen activator receptors, and matrix metalloproteinases. Thus, the fibrinolytic system linked by an activation cascade may lead to focal and transient degradation of tight junction proteins that allows B. burgdorferi to invade the CNS.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Borrelia burgdorferi/metabolismo , Enfermedad de Lyme/metabolismo , Péptido Hidrolasas/metabolismo , Barrera Hematoencefálica/microbiología , Células Endoteliales/metabolismo , Humanos , Enfermedad de Lyme/microbiología
20.
J Parasitol ; 90(5): 970-9, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15562595

RESUMEN

The neurological manifestations of sleeping sickness in man are attributed to the penetration of the blood-brain barrier (BBB) and invasion of the central nervous system by Trypanosoma brucei gambiense and Trypanosoma brucei rhodesiense. However, how African trypanosomes cross the BBB remains an unresolved issue. We have examined the traversal of African trypanosomes across the human BBB using an in vitro BBB model system constructed of human brain microvascular endothelial cells (BMECs) grown on Costar Transwell inserts. Human-infective T. b. gambiense strain IL 1852 was found to cross human BMECs far more readily than the animal-infective Trypanosoma brucei brucei strains 427 and TREU 927. Tsetse fly-infective procyclic trypomastigotes did not cross the human BMECs either alone or when coincubated with bloodstreamform T. b. gambiense. After overnight incubation, the integrity of the human BMEC monolayer measured by transendothelial electrical resistance was maintained on the inserts relative to the controls when the endothelial cells were incubated with T. b. brucei. However, decreases in electrical resistance were observed when the BMEC-coated inserts were incubated with T. b. gambiense. Light and electron microscopy studies revealed that T. b. gambiense initially bind at or near intercellular junctions before crossing the BBB paracellularly. This is the first demonstration of paracellular traversal of African trypanosomes across the BBB. Further studies are required to determine the mechanism of BBB traversal by these parasites at the cellular and molecular level.


Asunto(s)
Barrera Hematoencefálica/parasitología , Células Endoteliales/parasitología , Trypanosoma brucei brucei/fisiología , Trypanosoma brucei gambiense/fisiología , Animales , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/ultraestructura , Calcio/metabolismo , Línea Celular , Impedancia Eléctrica , Células Endoteliales/ultraestructura , Humanos , Técnicas In Vitro , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...