Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
J Neurol Sci ; 450: 120663, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37182424

ABSTRACT

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.


Subject(s)
Malaria, Cerebral , Meningitis , Child , Child, Preschool , Humans , Brain-Derived Neurotrophic Factor , Cytokines/cerebrospinal fluid , Interleukin-6/cerebrospinal fluid , Malaria, Cerebral/cerebrospinal fluid , Meningitis/cerebrospinal fluid
2.
Int J Mol Sci ; 24(8)2023 Apr 07.
Article in English | MEDLINE | ID: mdl-37108071

ABSTRACT

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.


Subject(s)
Astrocytes , Endothelial Protein C Receptor , Malaria, Cerebral , Humans , Astrocytes/metabolism , Brain/metabolism , Culture Media, Conditioned/metabolism , Endothelial Protein C Receptor/metabolism , Endothelium/metabolism , Oligodendroglia/metabolism
3.
Malar J ; 19(1): 266, 2020 Jul 23.
Article in English | MEDLINE | ID: mdl-32703204

ABSTRACT

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.


Subject(s)
Malaria, Cerebral , Malaria, Falciparum , Plasmodium falciparum/physiology , Erythrocytes/parasitology , Humans , Malaria, Cerebral/complications , Malaria, Cerebral/physiopathology , Malaria, Falciparum/complications , Malaria, Falciparum/physiopathology , Quality of Life
4.
Front Cell Neurosci ; 13: 405, 2019.
Article in English | MEDLINE | ID: mdl-31616251

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-30849122

ABSTRACT

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.


Subject(s)
Citrulline/pharmacology , Malaria, Cerebral/metabolism , Malaria, Cerebral/prevention & control , Animals , Arginase/blood , Arginine/administration & dosage , Arginine/blood , Arginine/deficiency , Blood-Brain Barrier/drug effects , Citrulline/administration & dosage , Dietary Supplements , Disease Models, Animal , Free Radical Scavengers/administration & dosage , Humans , Malaria, Cerebral/etiology , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Plasmodium berghei , Superoxides/metabolism , Urea/metabolism
6.
Fluids Barriers CNS ; 15(1): 12, 2018 Apr 23.
Article in English | MEDLINE | ID: mdl-29688865

ABSTRACT

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.


Subject(s)
Blood-Brain Barrier/physiopathology , Gray Matter/blood supply , Gray Matter/physiopathology , White Matter/blood supply , White Matter/physiopathology , Animals , Humans , In Vitro Techniques , Models, Biological , Neurovascular Coupling/physiology
7.
Thromb Haemost ; 114(5): 1038-48, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26155776

ABSTRACT

The Endothelial Protein C receptor (EPCR) is essential for the anticoagulant and cytoprotective functions of the Protein C (PC) system. Selected variants of the malaria parasite protein, Plasmodium falciparum Erythrocyte Membrane Protein 1 (PfEMP1) associated with severe malaria, including cerebral malaria, specifically target EPCR on vascular endothelial cells. Here, we examine the cellular response to PfEMP1 engagement to elucidate its role in malaria pathogenesis. Binding of the CIDRα1.1 domain of PfEMP1 to EPCR obstructed activated PC (APC) binding to EPCR and induced a loss of cellular EPCR functions. CIDRα1.1 severely impaired endothelial PC activation and effectively blocked APC-mediated activation of protease-activated receptor-1 (PAR1) and associated barrier protective effects of APC on endothelial cells. A soluble EPCR variant (E86A-sEPCR) bound CIDRα1.1 with high affinity and did not interfere with (A)PC binding to cellular EPCR. E86A-sEPCR used as a decoy to capture PfEMP1, permitted normal PC activation on endothelial cells, normal barrier protective effects of APC, and greatly reduced cytoadhesion of infected erythrocytes to brain endothelial cells. These data imply important contributions of PfEMP1-induced protein C pathway defects in the pathogenesis of severe malaria. Furthermore, the E86A-sEPCR decoy provides a proof-of-principle strategy for the development of novel adjunct therapies for severe malaria.


Subject(s)
Antigens, CD/metabolism , Endothelial Cells/physiology , Malaria/physiopathology , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Receptors, Cell Surface/metabolism , Antigens, CD/genetics , Brain/pathology , Cells, Cultured , Cytoprotection/drug effects , Endothelial Cells/drug effects , Endothelial Cells/parasitology , Endothelial Protein C Receptor , Humans , Malaria/drug therapy , Malaria/parasitology , Mutant Proteins/administration & dosage , Protein Binding/drug effects , Protein C/metabolism , Protein Structure, Tertiary/genetics , Protozoan Proteins/genetics , Receptor, PAR-1/metabolism , Receptors, Cell Surface/genetics , Signal Transduction/drug effects
8.
J Exp Med ; 212(9): 1391-403, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26216124

ABSTRACT

After being delivered by the bite from an infected mosquito, Plasmodium sporozoites enter the blood circulation and infect the liver. Previous evidence suggests that Kupffer cells, a macrophage-like component of the liver blood vessel lining, are traversed by sporozoites to initiate liver invasion. However, the molecular determinants of sporozoite-Kupffer cell interactions are unknown. Understanding the molecular basis for this specific recognition may lead to novel therapeutic strategies to control malaria. Using a phage display library screen, we identified a peptide, P39, that strongly binds to the Kupffer cell surface and, importantly, inhibits sporozoite Kupffer cell entry. Furthermore, we determined that P39 binds to CD68, a putative receptor for sporozoite invasion of Kupffer cells that acts as a gateway for malaria infection of the liver.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Kupffer Cells/metabolism , Malaria/metabolism , Plasmodium berghei/metabolism , Sporozoites/metabolism , Animals , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Kupffer Cells/parasitology , Kupffer Cells/pathology , Liver/metabolism , Liver/parasitology , Liver/pathology , Malaria/genetics , Malaria/pathology , Male , Mice , Mice, Knockout , Peptide Library , Rats , Rats, Sprague-Dawley
9.
Cell Microbiol ; 17(12): 1883-99, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26119044

ABSTRACT

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.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion , Endothelial Cells/physiology , Erythrocytes/parasitology , Host-Pathogen Interactions , Plasmodium falciparum/physiology , Receptors, Cell Surface/metabolism , Cells, Cultured , Endothelial Protein C Receptor , Humans , Ligation , Treatment Outcome
10.
Proc Natl Acad Sci U S A ; 109(26): E1782-90, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22619321

ABSTRACT

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.


Subject(s)
Brain/blood supply , Cell Adhesion , Endothelium, Vascular/pathology , Erythrocytes/parasitology , Genes, Protozoan , Malaria, Cerebral/parasitology , Plasmodium falciparum/physiology , Animals , Child, Preschool , Erythrocytes/pathology , Humans , Malaria, Cerebral/pathology , Plasmodium falciparum/genetics
11.
J Cereb Blood Flow Metab ; 32(6): 983-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22491155

ABSTRACT

Minocycline has been proposed as a way to blunt neurovascular injury from matrix metalloproteinases (MMPs) during stroke. However, recent clinical trials suggest that high levels of minocycline may have deleterious side-effects. Here, we showed that very high minocycline concentrations damage endothelial cells via calpain/caspase pathways. To alleviate this potential cytotoxicity, we encapsulated minocycline in liposomes. Low concentrations of minocycline could not reduce tumor necrosis factor α (TNFα)-induced MMP-9 release from endothelial cells. But low concentrations of minocycline-loaded liposomes significantly reduced TNFα-induced MMP-9 release. This study provides proof-of-concept that liposomes may be used to deliver lower levels of minocycline for targeting MMPs in cerebral endothelium.


Subject(s)
Anti-Bacterial Agents/pharmacology , Brain/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Minocycline/pharmacology , Brain/cytology , Cell Line , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Humans , Liposomes , Matrix Metalloproteinase 9/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
PLoS One ; 6(10): e24784, 2011.
Article in English | MEDLINE | ID: mdl-22043276

ABSTRACT

This study examined the ability of P.falciparum-infected erythrocytes (IE) to induce a pro-adhesive environment in the host endothelium during malaria infection, prior to the systemic cytokine activation seen in the later phase of disease. Previous work had shown increases in receptor levels but had not measured to actual impact on IE binding. Using a co-culture system with a range of endothelial cells (EC) and IE with different cytoadherent properties, we have characterised the specific expression of adhesion receptors and subsequent IE binding by FACS and adhesion assays. We have also examined the specific signalling pathways induced during co-culture that are potentially involved in the induction of receptor expression. The results confirmed that ICAM-1 is up-regulated, albeit at much lower levels than seen with TNF activation, in response to co-culture with infected erythrocytes in all three tissue endothelial cell types tested but that up-regulation of VCAM-1 is tissue-dependent. This small increase in the levels of EC receptors correlated with large changes in IE adhesion ability. Co-culture with either RBC or IE increased the potential of subsequent adhesion indicating priming/modulation effects on EC which make them more susceptible to adhesion and thereby the recruitment of IE. Trypsin surface digestion of IE and the use of a Pfsbp1-knockout (ko) parasite line abrogated the up-regulation of ICAM-1 and reduced IE binding to EC suggesting that PfEMP-1 and other molecules exported to the IE surface via the PfSBP1 pathway are major mediators of this phenotype. This was also supported by the higher induction of EC adhesion receptors by adherent IE compared to isogenic, non-adherent lines.


Subject(s)
Endothelial Cells/metabolism , Erythrocytes/parasitology , Gene Expression Regulation , Host-Parasite Interactions , Plasmodium falciparum/physiology , Cell Adhesion/genetics , Cell Adhesion Molecules/genetics , Coculture Techniques , Humans , Intercellular Adhesion Molecule-1/genetics
13.
Cell Microbiol ; 13(10): 1470-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21824246

ABSTRACT

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.


Subject(s)
Astrocytes/physiology , Blood-Brain Barrier/physiology , Endothelial Cells/physiology , Host-Pathogen Interactions , Neurons/physiology , Animals , Blood-Brain Barrier/microbiology , Blood-Brain Barrier/parasitology , Blood-Brain Barrier/virology , Endothelial Cells/microbiology , Endothelial Cells/parasitology , Endothelial Cells/virology , Humans
14.
Malar J ; 10: 23, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21288352

ABSTRACT

At the 2010 Keystone Symposium on "Malaria: new approaches to understanding Host-Parasite interactions", an extra scientific session to discuss animal models in malaria research was convened at the request of participants. This was prompted by the concern of investigators that skepticism in the malaria community about the use and relevance of animal models, particularly rodent models of severe malaria, has impacted on funding decisions and publication of research using animal models. Several speakers took the opportunity to demonstrate the similarities between findings in rodent models and human severe disease, as well as points of difference. The variety of malaria presentations in the different experimental models parallels the wide diversity of human malaria disease and, therefore, might be viewed as a strength. Many of the key features of human malaria can be replicated in a variety of nonhuman primate models, which are very under-utilized. The importance of animal models in the discovery of new anti-malarial drugs was emphasized. The major conclusions of the session were that experimental and human studies should be more closely linked so that they inform each other, and that there should be wider access to relevant clinical material.


Subject(s)
Disease Models, Animal , Malaria/immunology , Primates/immunology , Animal Experimentation , Animals , Humans , Immunity, Innate , Malaria/parasitology , Mice , Plasmodium/immunology , Plasmodium/physiology , Primates/parasitology
15.
Cerebrovasc Dis ; 30(3): 290-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20664263

ABSTRACT

BACKGROUND: Increasing evidence suggests that beyond its antiplatelet properties, dipyridamole may have pleiotropic effects on other cells within the neurovascular elements of the brain. In this experimental cellular study, we asked whether dipyridamole can ameliorate brain endothelial injury after exposure to inflammatory and metabolic insults. METHODS: Human brain endothelial cells were grown in culture, and exposed to TNFalpha (continuously for 20 h) or subjected to oxygen-glucose deprivation (OGD; 6 h of insult followed by 18 h recovery). Expression of ICAM-1, VCAM-1 and PECAM-1 were measured by immunoblotting. Matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9) in the conditioned media were quantified via zymography. MTT mitochondrial activity was measured to assess endothelial cell viability. RESULTS: Exposure of human brain endothelial cells to TNFalpha (12.5-50 ng/ml) induced a clear increase in protein levels of ICAM-1, VCAM-1 and MMP-9. TNFalpha did not alter PECAM-1. Dipyridamole (1-5 muM) significantly attenuated ICAM-1 and MMP-9 levels after this inflammatory insult. No significant effects of dipyridamole were noted for VCAM-1. Six-hour OGD induced moderate endothelial cell death accompanied by a release of MMP-9. Dipyridamole significantly decreased MMP-9 levels and cell death after this metabolic insult. CONCLUSIONS: These results suggest that dipyridamole may ameliorate brain endothelial injury after inflammation and/or metabolic insults. How these putative cellular mechanisms may relate to clinical outcomes and conditions in stroke patients remains to be elucidated.


Subject(s)
Brain/blood supply , Cytotoxins/pharmacology , Dipyridamole/therapeutic use , Endothelium, Vascular/drug effects , Inflammation/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , Tumor Necrosis Factor-alpha/pharmacology , Brain/metabolism , Cell Hypoxia/physiology , Cell Line , Cell Survival/drug effects , Dipyridamole/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Inflammation/metabolism , Intercellular Adhesion Molecule-1/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Phosphodiesterase Inhibitors/pharmacology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
16.
Blood ; 114(19): 4243-52, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19713460

ABSTRACT

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.


Subject(s)
Malaria, Cerebral/genetics , Malaria, Cerebral/physiopathology , Malaria, Falciparum/genetics , Malaria, Falciparum/physiopathology , NF-kappa B/physiology , Plasmodium falciparum/pathogenicity , Animals , Apoptosis , Blood-Brain Barrier , Brain/blood supply , Brain/parasitology , Cells, Cultured , Endothelial Cells/parasitology , Endothelial Cells/pathology , Endothelial Cells/physiology , Erythrocytes/parasitology , Erythrocytes/physiology , Genome-Wide Association Study , Humans , Immunity, Innate , Inflammation/genetics , Inflammation/parasitology , Inflammation/pathology , Inflammation/physiopathology , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Models, Biological , Oligonucleotide Array Sequence Analysis , Signal Transduction
17.
Stroke ; 40(2): 652-5, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18974377

ABSTRACT

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.


Subject(s)
Astrocytes/metabolism , Brain Chemistry/drug effects , Endothelial Cells/metabolism , Lithium Chloride/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Astrocytes/drug effects , Blotting, Western , Brain/cytology , Brain-Derived Neurotrophic Factor/biosynthesis , Chromones/pharmacology , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/biosynthesis , Humans , Indoles/pharmacology , Maleimides/pharmacology , Morpholines/pharmacology , Up-Regulation/drug effects
18.
Stroke ; 39(9): 2538-43, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18635843

ABSTRACT

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.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Brain Edema/enzymology , Brain Infarction/enzymology , Brain Ischemia/enzymology , Flavanones/therapeutic use , Ischemic Attack, Transient/enzymology , Animals , Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/drug effects , Arachidonate 15-Lipoxygenase/genetics , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Edema/physiopathology , Brain Edema/prevention & control , Brain Infarction/drug therapy , Brain Infarction/physiopathology , Brain Ischemia/drug therapy , Brain Ischemia/physiopathology , Cells, Cultured , Claudin-5 , Cytoprotection/drug effects , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Hydrogen Peroxide/pharmacology , Immunoglobulin G/metabolism , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/physiopathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Oxidative Stress/physiology , Tight Junctions/drug effects , Tight Junctions/metabolism
19.
Proc Natl Acad Sci U S A ; 105(21): 7582-7, 2008 May 27.
Article in English | MEDLINE | ID: mdl-18495934

ABSTRACT

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.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Brain/cytology , Brain/metabolism , Cell Communication , Endothelium, Vascular/physiology , Neurons/physiology , Animals , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Integrin beta1/metabolism , Male , Mice , Mice, Inbred Strains , Neurons/cytology , Neurons/metabolism , Oxidative Stress , Protein Serine-Threonine Kinases/metabolism
20.
Antimicrob Agents Chemother ; 51(12): 4471-3, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17875991

ABSTRACT

Cycloheximide, ketoconazole, or preexposure of organisms to cytochalasin D prevented Balamuthia mandrillaris-associated cytopathogenicity in human brain microvascular endothelial cells, which constitute the blood-brain barrier. In an assay for inhibition of cyst production, these three agents prevented the production of cysts, suggesting that the biosynthesis of proteins and ergosterol and the polymerization of actin are important in cytopathogenicity and encystment.


Subject(s)
Amebicides/pharmacology , Amoeba/drug effects , Endothelial Cells/drug effects , Actins/metabolism , Amoeba/growth & development , Amoeba/metabolism , Animals , Cell Line , Cell Survival/drug effects , Cycloheximide/pharmacology , Cytochalasin D/pharmacology , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/parasitology , Ergosterol/biosynthesis , Humans , Ketoconazole/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...