Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Neuropathol Appl Neurobiol ; 44(3): 328-340, 2018 04.
Article in English | MEDLINE | ID: mdl-28453876

ABSTRACT

AIMS: While vascular pathology is a common feature of a range of neurodegenerative diseases, we hypothesized that vascular changes occur in association with normal ageing. Therefore, we aimed to characterize age-associated changes in the blood-brain barrier (BBB) in human and mouse cohorts. METHODS: Immunohistochemistry and Evans blue assays were used to characterize BBB dysfunction (tight junction protein expression and serum plasma protein accumulation), vascular pathology (pericyte loss and vascular density) and glial pathology (astrocyte and microglial density) in ageing neurological control human prefrontal cortex (a total of 23 cases from 5 age groups representing the spectrum of young adult to old age: 20-30 years, 31-45 years, 46-60 years, 61-75 years and 75+) and C57BL/6 mice (3 months, 12 months, 18 months and 24 months, n = 5/6 per group). RESULTS: Quantification of the tight junction protein ZO-1 within the cortex and cerebellum of the mouse cohort showed a significant trend to both increased number (cortex P < 0.001, cerebellum P < 0.001) and length (cortex P < 0.001, cerebellum P < 0.001) of junctional breaks associated with increasing age. GFAP expression significantly correlated with ageing in the mice (P = 0.037). In the human cohort, assessment of human protein accumulation (albumin, fibrinogen and human IgG) demonstrated cells morphologically resembling clasmatodendritic astrocytes, indicative of BBB dysfunction. Semiquantitative assessment of astrogliosis in the cortex expression revealed an association with age (P = 0.003), while no age-associated changes in microglial pathology, microvascular density or pericyte coverage were detected. CONCLUSIONS: This study demonstrates BBB dysfunction in normal brain ageing, both in human and mouse cohorts.


Subject(s)
Aging/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Tight Junctions/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Animals , Astrocytes/metabolism , Female , Humans , Male , Mice , Middle Aged , Pericytes/metabolism , Young Adult , Zonula Occludens-1 Protein/metabolism
2.
BMC Microbiol ; 15: 211, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26467858

ABSTRACT

BACKGROUND: Neonatal meningitis-causing Escherichia coli (NMEC) is the predominant Gram-negative bacterial pathogen associated with meningitis in newborn infants. High levels of heterogeneity and diversity have been observed in the repertoire of virulence traits and other characteristics among strains of NMEC making it difficult to define the NMEC pathotype. The objective of the present study was to identify genotypic and phenotypic characteristics of NMEC that can be used to distinguish them from commensal E. coli. METHODS: A total of 53 isolates of NMEC obtained from neonates with meningitis and 48 isolates of fecal E. coli obtained from healthy individuals (HFEC) were comparatively evaluated using five phenotypic (serotyping, serum bactericidal assay, biofilm assay, antimicorbial susceptibility testing, and in vitro cell invasion assay) and three genotypic (phylogrouping, virulence genotyping, and pulsed-field gel electrophoresis) methods. RESULTS: A majority (67.92%) of NMEC belonged to B2 phylogenetic group whereas 59% of HFEC belonged to groups A and D. Serotyping revealed that the most common O and H types present in NMEC tested were O1 (15%), O8 (11.3%), O18 (13.2%), and H7 (25.3%). In contrast, none of the HFEC tested belonged to O1 or O18 serogroups. The most common serogroup identified in HFEC was O8 (6.25%). The virulence genotyping reflected that more than 70% of NMEC carried kpsII, K1, neuC, iucC, sitA, and vat genes with only less than 27% of HFEC possessing these genes. All NMEC and 79% of HFEC tested were able to invade human cerebral microvascular endothelial cells. No statistically significant difference was observed in the serum resistance phenotype between NMEC and HFEC. The NMEC strains demonstrated a greater ability to form biofilms in Luria Bertani broth medium than did HFEC (79.2% vs 39.9%). CONCLUSION: The results of our study demonstrated that virulence genotyping and phylogrouping may assist in defining the potential NMEC pathotype.


Subject(s)
Escherichia coli Infections/microbiology , Escherichia coli/classification , Escherichia coli/isolation & purification , Genotype , Meningitis, Bacterial/microbiology , Phenotype , Biofilms/growth & development , Blood Bactericidal Activity , Endocytosis , Escherichia coli/genetics , Escherichia coli/physiology , Humans , Infant, Newborn , Microbial Sensitivity Tests , Molecular Typing , Serogroup , Virulence Factors/genetics
3.
Br J Pharmacol ; 172(22): 5390-402, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26362823

ABSTRACT

BACKGROUND AND PURPOSE: Stroke and Alzheimer's disease (AD) are related pathologies in which the cerebrovascular system is involved. Plasma levels of semicarbazide-sensitive amine oxidase/vascular adhesion protein 1 (SSAO/VAP-1, also known as Primary Amine Oxidase -PrAO) are increased in both stroke and AD patients and contribute to the vascular damage. During inflammation, its enzymatic activity mediates leukocyte recruitment to the injured tissue, inducing damage in the blood-brain barrier (BBB) and neuronal tissue. We hypothesized that by altering cerebrovascular function, SSAO/VAP-1 might play a role in the stroke-AD transition. Therefore, we evaluated the protective effect of the novel multitarget-directed ligand DPH-4, initially designed for AD therapy, on the BBB. EXPERIMENTAL APPROACH: A human microvascular brain endothelial cell line expressing human SSAO/VAP-1 was generated, as the expression of SSAO/VAP-1 is lost in cultured cells. To simulate ischaemic damage, these cells were subjected to oxygen and glucose deprivation (OGD) and re-oxygenation conditions. The protective role of DPH-4 was then evaluated in the presence of methylamine, an SSAO substrate, and/or ß-amyloid (Aß). KEY RESULTS: Under our conditions, DPH-4 protected brain endothelial cells from OGD and re-oxygenation-induced damage, and also decreased SSAO-dependent leukocyte adhesion. DPH-4 was also effective at preventing the damage induced by OGD and re-oxygenation in the presence of Aß as a model of AD pathology. CONCLUSIONS AND IMPLICATIONS: From these results, we concluded that the multitarget compound DPH-4 might be of therapeutic benefit to delay the onset and/or progression of the neurological pathologies associated with stroke and AD, which appear to be linked.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Brain Ischemia/metabolism , Cell Adhesion Molecules/metabolism , Hydroxyquinolines/pharmacology , Neuroprotective Agents/pharmacology , Piperidines/pharmacology , Cell Hypoxia/physiology , Cell Line , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glucose/metabolism , Humans , Microvessels/cytology , Oxygen/metabolism
4.
J Mater Chem B ; 3(46): 9053-9059, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-32263036

ABSTRACT

Herein, we report the synthesis and characterization of nanospheres of a biodegradable zinc-imidazolate polymers (ZIPs) as a proof-of-concept delivery vehicle into human brain endothelial cells, the main component of the blood-brain barrier (BBB). The ZIP particles can readily encapsulate functional molecules such as fluorophores and inorganic nanoparticles at the point of synthesis producing stable colloidal dispersions. Our results show that these biodegradable particles are not cytotoxic, and are able to penetrate and release cargo species to human brain capillary endothelial cells in vitro thus exhibiting significant potential as a novel platform for brain targeting treatments.

5.
Biomed Microdevices ; 15(1): 145-50, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22955726

ABSTRACT

The blood-brain barrier (BBB) is a unique feature of the human body, preserving brain homeostasis and preventing toxic substances to enter the brain. However, in various neurodegenerative diseases, the function of the BBB is disturbed. Mechanisms of the breakdown of the BBB are incompletely understood and therefore a realistic model of the BBB is essential. We present here the smallest model of the BBB yet, using a microfluidic chip, and the immortalized human brain endothelial cell line hCMEC/D3. Barrier function is modulated both mechanically, by exposure to fluid shear stress, and biochemically, by stimulation with tumor necrosis factor alpha (TNF-α), in one single device. The device has integrated electrodes to analyze barrier tightness by measuring the transendothelial electrical resistance (TEER). We demonstrate that hCMEC/D3 cells could be cultured in the microfluidic device up to 7 days, and that these cultures showed comparable TEER values with the well-established Transwell assay, with an average (± SEM) of 36.9 Ω.cm(2) (± 0.9 Ω.cm(2)) and 28.2 Ω.cm(2) (± 1.3 Ω.cm(2)) respectively. Moreover, hCMEC/D3 cells on chip expressed the tight junction protein Zonula Occludens-1 (ZO-1) at day 4. Furthermore, shear stress positively influenced barrier tightness and increased TEER values with a factor 3, up to 120 Ω.cm(2). Subsequent addition of TNF-α decreased the TEER with a factor of 10, down to 12 Ω.cm(2). This realistic microfluidic platform of the BBB is very well suited to study barrier function in detail and evaluate drug passage to finally gain more insight into the treatment of neurodegenerative diseases.


Subject(s)
Blood-Brain Barrier/metabolism , Mechanical Phenomena , Microfluidic Analytical Techniques/instrumentation , Biomechanical Phenomena , Blood-Brain Barrier/cytology , Cell Line , Electric Impedance , Endothelial Cells/metabolism , Humans , Microscopy, Confocal
6.
Brain Res ; 1437: 115-26, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22226505

ABSTRACT

Chemokines are low relative molecular mass proteins, which have chemoattractant actions on many cell types. The chemokine, CCL2, has been shown to play a major role in the recruitment of monocytes in central nervous system (CNS) lesions in multiple sclerosis (MS). Since resident astrocytes constitute a major source of chemokine synthesis including CCL2, we were interested to assess the regulation of CCL2 by astrocytes. We showed that CCL2 bound to the cell surface of astrocytes and binding was not modulated by inflammatory conditions. However, CCR2 protein was not detected nor was activation of the classical CCR2 downstream signaling pathways. Recent studies have shown that non-signaling decoy chemokine receptors bind and modulate the expression of chemokines at site of inflammation. Here, we show that the D6 chemokine decoy receptor is constitutively expressed by primary human adult astrocytes at both mRNA and protein level. In addition, CCL3, which binds to D6, but not CCL19, which does not bind to D6, displaced CCL2 binding to astrocytes; indicating that CCL2 may bind to this cell type via the D6 receptor. Our results suggest that CCL2 binding to primary adult human astrocytes is CCR2-independent and is likely to be mediated via the D6 decoy chemokine receptor. Therefore we propose that astrocytes are implicated in both the establishment of chemokine gradients for the migration of leukocytes into and within the CNS and in the regulation of CCL2 levels at inflammatory sites in the CNS.


Subject(s)
Astrocytes/metabolism , Chemokine CCL2/metabolism , Receptors, CCR2/metabolism , Adult , Astrocytes/pathology , Cells, Cultured , Humans , Inflammation Mediators/metabolism , Protein Binding/physiology , Receptors, CCR10/metabolism , Receptors, CCR10/physiology , Chemokine Receptor D6
7.
FASEB J ; 24(1): 229-41, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19770225

ABSTRACT

Cerebral amyloid angiopathy (CAA) is an age-associated condition and a common finding in Alzheimer's disease in which amyloid-beta (Abeta) vascular deposits are featured in >80% of the cases. Familial Abeta variants bearing substitutions at positions 21-23 are primarily associated with CAA, although they manifest with strikingly different clinical phenotypes: cerebral hemorrhage or dementia. The recently reported Piedmont L34V Abeta mutant, located outside the hot spot 21-23, shows a similar hemorrhagic phenotype, albeit less aggressive than the widely studied Dutch E22Q variant. We monitored the apoptotic events occurring after stimulation of human brain microvascular endothelial and smooth muscle cells with nonfibrillar structures of both variants and wild-type Abeta40. Induction of analogous caspase-mediated mitochondrial pathways was elicited by all peptides, although within different time frames and intensity. Activated pathways were susceptible to pharmacological modulation either through direct inhibition of mitochondrial cytochrome c release or by the action of pan- and pathway-specific caspase inhibitors, giving a clear indication of the independent or synergistic engagement of both extrinsic and intrinsic mechanisms. Structural analyses of the Abeta peptides showed that apoptosis preceded fibril formation, correlating with the presence of oligomers and/or protofibrils. The data support the notion that rare genetic mutations constitute unique paradigms to understand the molecular pathogenesis of CAA.


Subject(s)
Amyloid beta-Peptides/genetics , Brain/blood supply , Cerebral Amyloid Angiopathy, Familial/genetics , Cerebral Amyloid Angiopathy, Familial/pathology , Amino Acid Substitution , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Apoptosis , Brain/metabolism , Brain/pathology , Caspases/metabolism , Cell Line , Cerebral Amyloid Angiopathy, Familial/metabolism , Cytochromes c/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Genetic Variation , Humans , Mitochondria/metabolism , Mutation , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
8.
J Cell Mol Med ; 14(5): 1101-12, 2010 May.
Article in English | MEDLINE | ID: mdl-19438816

ABSTRACT

Vascular dysfunction is emerging as a key pathological hallmark in Alzheimer's disease (AD). A leaky blood-brain barrier (BBB) has been described in AD patient tissue and in vivo AD mouse models. Brain endothelial cells (BECs) are linked together by tight junctional (TJ) proteins, which are a key determinant in restricting the permeability of the BBB. The amyloid beta (Abeta) peptides of 1-40 and 1-42 amino acids are believed to be pivotal in AD pathogenesis. We therefore decided to investigate the effect of Abeta 1-40, the Abeta variant found at the highest concentration in human plasma, on the permeability of an immortalized human BEC line, hCMEC/D3. Abeta 1-40 induced a marked increase in hCMEC/D3 cell permeability to the paracellular tracer 70 kD FITC-dextran when compared with cells incubated with the scrambled Abeta 1-40 peptide. Increased permeability was associated with a specific decrease, both at the protein and mRNA level, in the TJ protein occludin, whereas claudin-5 and ZO-1 were unaffected. JNK and p38MAPK inhibition prevented both Abeta 1-40-mediated down-regulation of occludin and the increase in paracellular permeability in hCMEC/D3 cells. Our findings suggest that the JNK and p38MAPK pathways might represent attractive therapeutic targets for preventing BBB dysfunction in AD.


Subject(s)
Amyloid beta-Peptides/pharmacology , Brain/cytology , Cell Membrane Permeability/drug effects , Down-Regulation/drug effects , Endothelial Cells/enzymology , Membrane Proteins/genetics , Mitogen-Activated Protein Kinases/metabolism , Cell Survival/drug effects , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Membrane Proteins/metabolism , Occludin , Peptides/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
9.
Cytogenet Genome Res ; 126(4): 313-7, 2009.
Article in English | MEDLINE | ID: mdl-19864871

ABSTRACT

The immortalized human cerebral microvessel endothelial cell line hCMEC/D3 has been repeatedly used as a model of human blood-brain barrier (BBB). hCMEC/D3 cells between passage 25 and 35 are most often applied in research, remained phenotypically nontransformed, and cells maintained many characteristics of human brain endothelial cells. Also hCMEC/D3 was thought to have conserved a normal diploid karyotype over all these passages. Here we characterized the cell line using high-resolution multicolor fluorescence in situ hybridization (FISH) approaches and revealed a complex karyotype in the 30th passage. Clonal cryptic unbalanced structural rearrangements and numerical aberrations were discovered and described as follows: 45 approximately 48,XX, -X,del(5)(q11)[2],del(9)(q11)[3],+9[3],del(11)(q13 approximately 14)[2], der(14)t(14;21)(q32.33;q22.3)[28],der(15)t(9;15)(p11;p11)[13], dup(15)(p11q11)[5],der(21)t(17;21)(p12;q22)[9],-22[6][cp28]. In summary, a complex karyotype with clonal unbalanced chromosomal rearrangements is present in hCMEC/D3. Thus, we solicit to include molecular cytogenetics in the testing of all cell lines prior to application of their use in complex studies.


Subject(s)
Brain/blood supply , Brain/cytology , Endothelial Cells/cytology , Microvessels/cytology , Cell Line , Chromosome Aberrations , Humans , Karyotyping
10.
FASEB J ; 19(13): 1872-4, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16141364

ABSTRACT

Establishment of a human model of the blood-brain barrier has proven to be a difficult goal. To accomplish this, normal human brain endothelial cells were transduced by lentiviral vectors incorporating human telomerase or SV40 T antigen. Among the many stable immortalized clones obtained by sequential limiting dilution cloning of the transduced cells, one was selected for expression of normal endothelial markers, including CD31, VE cadherin, and von Willebrand factor. This cell line, termed hCMEC/D3, showed a stable normal karyotype, maintained contact-inhibited monolayers in tissue culture, exhibited robust proliferation in response to endothelial growth factors, and formed capillary tubes in matrix but no colonies in soft agar. hCMEC/D3 cells expressed telomerase and grew indefinitely without phenotypic dedifferentiation. These cells expressed chemokine receptors, up-regulated adhesion molecules in response to inflammatory cytokines, and demonstrated blood-brain barrier characteristics, including tight junctional proteins and the capacity to actively exclude drugs. hCMEC/D3 are excellent candidates for studies of blood-brain barrier function, the responses of brain endothelium to inflammatory and infectious stimuli, and the interaction of brain endothelium with lymphocytes or tumor cells. Thus, hCMEC/D3 represents the first stable, fully characterized, well-differentiated human brain endothelial cell line and should serve as a widely usable research tool.


Subject(s)
Blood-Brain Barrier , Brain/cytology , Brain/drug effects , Cell Culture Techniques/methods , Drug Resistance, Multiple , Endothelial Cells/cytology , Agar/chemistry , Animals , Antigens, CD , Antigens, Polyomavirus Transforming/biosynthesis , Antigens, Polyomavirus Transforming/genetics , Blood-Brain Barrier/drug effects , Blotting, Western , Brain/metabolism , Brain/pathology , Cadherins/biosynthesis , Capillaries/pathology , Cattle , Cell Adhesion , Cell Line , Cell Proliferation , Cells, Cultured , Cloning, Molecular , Collagen/pharmacology , Cytokines/metabolism , Drug Combinations , Endothelial Cells/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Female , Flow Cytometry , Humans , Immunohistochemistry , Karyotyping , Laminin/pharmacology , Lentivirus/genetics , Lymphocytes/metabolism , Microscopy, Fluorescence , Models, Biological , Perfusion , Permeability , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Proteoglycans/pharmacology , RNA/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Telomerase/genetics , Telomerase/metabolism , Time Factors , Up-Regulation , von Willebrand Factor/biosynthesis
11.
Arterioscler Thromb Vasc Biol ; 21(5): 810-6, 2001 May.
Article in English | MEDLINE | ID: mdl-11348879

ABSTRACT

Intercellular adhesion molecule (ICAM)-1 plays a vital role in the process of leukocyte transmigration through endothelial cell (EC) barriers and has been shown to mediate signal transduction events in ECs induced either by its cross-linking or by the binding of T lymphocytes. Immunoblotting of ICAM-1 of Triton X-100 detergent fractions demonstrated that the majority of ICAM-1 was contained within the detergent-soluble fraction (noncytoskeletal associated) under basal conditions. After cross-linking of endothelial ICAM-1 with monoclonal antibody or coculture with T lymphocytes, EC ICAM-1 was observed to partition with a Triton X-100-insoluble (cytoskeletal associated) fraction in a dose- and time-dependent manner. Redistribution of ICAM-1 was specific, inasmuch as no association with the Triton X-100-insoluble fraction was observed after cross-linking of vascular cell adhesion molecule-1, nor did cross-linking of ICAM-1 result in a redistribution of the platelet and EC adhesion molecule. ICAM-1 association with the endothelial cytoskeleton after cross-linking was unaffected after treatment of the cells with cytochalasin D, C3-transferase, removal of extracellular calcium ions, or chelation of intracellular calcium ions. These data show that ICAM-1 colocalizes with the endothelial cytoskeleton and associates with a detergent-insoluble fraction after cross-linking.


Subject(s)
Brain/metabolism , Cytoskeleton/metabolism , Endothelium, Vascular/metabolism , Intercellular Adhesion Molecule-1/metabolism , Actins/metabolism , Animals , Antibodies, Monoclonal/immunology , Calcium/metabolism , Caveolin 1 , Caveolins/metabolism , Cell Line , Cells, Cultured , Centrifugation, Density Gradient , Endothelium, Vascular/drug effects , Intercellular Adhesion Molecule-1/immunology , Octoxynol/chemistry , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rats , Solubility , Subcellular Fractions/metabolism , T-Lymphocytes/metabolism , Vascular Cell Adhesion Molecule-1/immunology , rhoA GTP-Binding Protein/metabolism
12.
Mol Cell Endocrinol ; 175(1-2): 81-92, 2001 Apr 25.
Article in English | MEDLINE | ID: mdl-11325518

ABSTRACT

In addition to their role in inflammation, cytokines like TNFalpha have been reported to regulate the adipose tissue function suggesting a role for these soluble mediators in metabolism. However, it is not known whether adipocytes have the capacity to secrete chemokines, a group of low molecular weight inflammatory mediators that control leukocyte migration into tissues. Here we show that primary cultures of human preadipocytes constitutively produce three chemokines, interleukin-8 (IL-8), macrophage inflammatory protein-1alpha (MIP-1alpha) and monocyte chemotactic protein-1 (MCP-1), while their level of expression is low in mature adipocytes. Upon TNFalpha treatment, the expression of all the three chemokines is upregulated in adipocytes differentiated in vitro. In addition, we describe the presence of seven different chemokine receptors, mainly in mature adipocytes, both in vitro and in human fat tissue sections. Prolonged stimulation of cultured human adipocytes with exogenous chemokines leads to a decrease in lipid content in association with the downregulation of PPARgamma mRNA expression. Moreover, chemokines positively control the secretion of leptin, a hormone that regulates appetite, by a post-transcriptional mechanism. These findings reveal a new role for chemokines in the regulation of adipose tissue and suggest a novel therapeutic basis for the treatment of obesity, diabetes and cachexia.


Subject(s)
Adipocytes/drug effects , Chemokines/pharmacology , Leptin/metabolism , Triglycerides/metabolism , Adipocytes/chemistry , Adipocytes/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL3 , Chemokine CCL4 , Chemokines/genetics , Chemokines/metabolism , Humans , Immunohistochemistry , Interleukin-8/genetics , Interleukin-8/metabolism , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/metabolism , RNA, Messenger/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Receptors, Leptin , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
13.
J Immunol ; 165(3): 1573-81, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10903766

ABSTRACT

Annexin 1 (ANX1), a calcium-binding protein, participates in the regulation of early inflammatory responses. Whereas some of its effects depend on intracellular interactions, a growing number of observations indicate that ANX1 may also act via autocrine/paracrine functions following externalization to the outer side of the plasma membrane. We studied the effects of ANX1 on leukocyte adhesion to endothelial cells using as a model system the monocytic cell line U937 and human bone marrow microvascular endothelial cells. Exogenous rANX1, as well as endogenous ANX1 externalized by U937 differentiated in vitro, inhibited monocyte firm adhesion to vascular endothelium. Both binding of ANX1 to U937 cells and ANX1-mediated inhibition of cell adhesion involved the short N-terminal domain of the ANX1 molecule. Under experimental conditions in which ANX1 inhibited U937 adhesion to human bone marrow microvascular endothelial cells, this protein specifically colocalized with the alpha 4 integrin, and a direct interaction between ANX1 and the alpha 4 integrin could be documented by immunoprecipitation experiments. Moreover, ANX1 competed with the endothelial integrin counterreceptor, VCAM-1, for binding to alpha 4 integrin. These results indicate that ANX1 plays an important physiological role in modulating monocyte firm adhesion to the endothelium.


Subject(s)
Annexin A1/immunology , Annexin A1/metabolism , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Integrins/physiology , Monocytes/immunology , Monocytes/metabolism , Receptors, Lymphocyte Homing/physiology , Autocrine Communication/immunology , Binding Sites/immunology , Binding, Competitive/immunology , Cell Adhesion/immunology , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Integrin alpha4beta1 , Integrins/metabolism , Ligands , Lymphocyte Function-Associated Antigen-1/metabolism , Microcirculation/cytology , Microcirculation/immunology , Microcirculation/metabolism , Paracrine Communication/immunology , Protein Binding/immunology , Receptors, Lymphocyte Homing/metabolism , U937 Cells , Vascular Cell Adhesion Molecule-1/metabolism
14.
J Virol ; 74(13): 6021-30, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10846084

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is associated with a variety of clinical manifestations, including tropical spastic paraparesis or HTLV-1-associated myelopathy (TSP/HAM). Viral detection in the central nervous system (CNS) of TSP/HAM patients demonstrates the ability of HTLV-1 to cross the blood-brain barrier (BBB). To investigate viral entry into the CNS, rat brain capillary endothelial cells were exposed to human lymphocytes chronically infected by HTLV-1 (MT2), to lymphocytes isolated from a seropositive patient, or to a control lymphoblastoid cell line (CEM). An enhanced adhesion to and migration through brain endothelial cells in vitro was observed with HTLV-1-infected lymphocytes. HTLV-1-infected lymphocytes also induced a twofold increase in the paracellular permeability of the endothelial monolayer. These effects were associated with an increased production of tumor necrosis factor alpha by HTLV-1-infected lymphocytes in the presence of brain endothelial cells. Ultrastructural analysis showed that contact between endothelial cells and HTLV-1-infected lymphocytes resulted in a massive and rapid budding of virions from lymphocytes, followed by their internalization into vesicles by brain endothelial cells and apparent release onto the basolateral side, suggesting that viral particles may cross the BBB using the transcytotic pathway. Our study also demonstrates that cell-cell fusion occurs between HTLV-1-infected lymphocytes and brain endothelial cells, with the latter being susceptible to transient HTLV-1 infection. These aspects may help us to understand the pathogenic mechanisms associated with neurological diseases induced by HTLV-1 infection.


Subject(s)
Brain/blood supply , Endothelium, Vascular/virology , Human T-lymphotropic virus 1/physiology , Animals , Brain/virology , Capillaries/cytology , Cell Adhesion/physiology , Cell Fusion , Cell Membrane Permeability , Cell Movement/physiology , Cells, Cultured , Central Nervous System/blood supply , Central Nervous System/virology , Coculture Techniques , Endocytosis , Endothelium, Vascular/cytology , Humans , Integrin alpha4beta1 , Integrins/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Rats , Receptors, Lymphocyte Homing/metabolism , T-Lymphocytes/physiology , T-Lymphocytes/virology , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Virion
15.
J Neurochem ; 73(5): 1954-63, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10537053

ABSTRACT

The blood-brain barrier (BBB) plays an important role in controlling the passage of molecules from the blood to the extracellular fluid environment of the brain. The multidrug efflux pump P-glycoprotein (P-gp) is highly expressed in the luminal membrane of brain capillary endothelial cells, thus forming a functional barrier to lipid-soluble drugs, notably, antitumor agents. It is of interest to develop an in vitro BBB model that stably expresses P-gp to investigate the mechanisms of regulation in expression and activity. The rat brain endothelial cell line, GPNT, was derived from a previously characterized rat brain endothelial cell line. A strong expression of P-gp was found in GPNT monocultures, whereas the multidrug resistance-associated pump Mrp1 was not expressed. The transendothelial permeability coefficient of the P-gp substrate vincristine across GPNT monolayers was close to the permeability coefficient of bovine brain endothelial cells cocultured with astrocytes, a previously documented in vitro BBB model. Furthermore, the P-gp blocker cyclosporin A induced a large increase in apical to basal permeability of vincristine. Thus, P-gp is highly functional in GPNT cells. A 1-h treatment of GPNT cells with dexamethasone resulted in decreased uptake of vincristine without any increase in P-gp expression. This effect could be mimicked by protein kinase C (PKC) activation and prevented by PKC inhibition, strongly suggesting that activation of P-gp function may involve a PKC-dependent pathway. These results document the GPNT cell line as a valuable in vitro model for studying drug transport and P-gp function at the BBB and suggest that activation of P-gp activity at the BBB might be considered in chemotherapeutic treatment of cancer patients.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/blood supply , Dexamethasone/pharmacology , Endothelium, Vascular/metabolism , Glucocorticoids/pharmacology , Multidrug Resistance-Associated Proteins , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Astrocytes/metabolism , Blood-Brain Barrier/physiology , Cattle , Cell Line, Transformed , Cell Membrane Permeability , Coculture Techniques , Colchicine/metabolism , Cyclosporine/pharmacology , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , MutS Homolog 3 Protein , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Vincristine/metabolism
16.
Free Radic Biol Med ; 27(5-6): 667-72, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10490287

ABSTRACT

In the present study, we investigated the changes in blood-brain barrier (BBB) permeability following brain endothelial cell exposure to different xenobiotics able to promote free radical generation during their metabolism. Our in vitro BBB model consisted of confluent monolayers of immortalized rat brain capillary endothelial cells (RBE4) grown on collagen-coated filters in the presence of C6 glioma cells grown in the lower compartment. We have recently shown that a range of xenobiotics, including menadione, nitrofurazone, and methylviologen (paraquat) may undergo monoelectronic redox cycling in isolated brain capillaries, giving rise to reactive oxygen species. In this study, addition of 100 microM menadione to the culture medium for 30 min significantly increased the permeability of endothelial cell monolayers to radiolabeled sucrose. The effect on endothelial permeability induced by menadione was dose-dependent and reversible. These permeability changes preceded the onset of cell death, as assessed by the Trypan blue exclusion method. Pre-incubation with superoxide dismutase and catalase blocked changes in sucrose permeability to control levels in a dose-dependent manner, suggesting the involvement of reactive oxygen species in menadione-induced BBB opening.


Subject(s)
Blood-Brain Barrier/drug effects , Cell Membrane Permeability/drug effects , Vitamin K/pharmacology , Animals , Benzoquinones/pharmacology , Brain/blood supply , Catalase/metabolism , Cell Line , Cell Survival/drug effects , Coculture Techniques , Endothelium, Vascular/drug effects , Free Radicals/pharmacology , Nitrofurazone/pharmacology , Paraquat/pharmacology , Rats , Sucrose/metabolism , Superoxide Dismutase/metabolism , Superoxides/pharmacology , Trypan Blue , Tumor Cells, Cultured
17.
Neurosci Lett ; 242(2): 105-8, 1998 Feb 13.
Article in English | MEDLINE | ID: mdl-9533405

ABSTRACT

The effect of a truncated form of the neurotoxin beta-amyloid peptide (A beta25-35) on rat brain vascular endothelial cells (RBE4 cells) was studied in cell culture. Toxic effects of the peptide were seen at 200 microg/ml A beta using a mitochondrial dehydrogenase activity (MTT) reduction assay, lactate dehydrogenase release and glucose consumption. Cell damage could be prevented completely at 200 microg/ml A beta and partially at 300 microg/ml A beta, by the dipeptide carnosine. Carnosine is a naturally occurring dipeptide found at high levels in brain tissue and innervated muscle of mammals including humans. Agents which share properties similar to carnosine, such as beta-alanine, homocarnosine, the anti-glycating agent aminoguanidine, and the antioxidant superoxide dismutase (SOD), also partially rescued cells, although not as effectively as carnosine. We postulate that the mechanism of carnosine protection lies in its anti-glycating and antioxidant activities, both of which are implicated in neuronal and endothelial cell damage during Alzheimer's disease. Carnosine may therefore be a useful therapeutic agent.


Subject(s)
Amyloid beta-Peptides/pharmacology , Carnosine/analogs & derivatives , Endothelium, Vascular/drug effects , Peptide Fragments/pharmacology , beta-Alanine/pharmacology , Animals , Carnosine/pharmacology , Cell Line, Transformed , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , L-Lactate Dehydrogenase/metabolism , Neuroprotective Agents/pharmacology , Rats , Superoxide Dismutase/pharmacology
18.
J Neurochem ; 70(2): 778-85, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9453574

ABSTRACT

The role of nonproductive infection of astrocytes by human immunodeficiency virus type 1 (HIV-1), characterized by the overexpression of nef, in brain disease progression is largely unknown. We investigated the consequences of stable expression of nef from the HIV-1 strain LAI in the human astrocytic cell line U373. DNA synthesis induced by endothelin-1 (ET-1) was largely decreased by nef. Stable expression of nef did not affect the ET-1-induced tyrosine phosphorylation of focal adhesion kinase, an adhesion-dependent pathway known to participate in DNA synthesis in astrocytes. Conversely, the activation of extracellular signal-regulated kinase (ERK) by ET-1 was largely inhibited in cells stably or transiently expressing nef. A similar inhibitory action of nef on ERK activation was observed after direct stimulation of G proteins. Furthermore, the inhibitory action of nef did not require protein kinase C (PKC) and affected mainly the PKC-independent pathway of ERK activation. Following chemokine receptor CXCR4-mediated infection of U373 cells stably expressing CXCR4 with the T-tropic HIV-1 strain m7-NDK, ET-1-induced activation of ERK was also inhibited. Altogether, these results indicate that intracellular signaling pathways associated with the growth factor activity of ET-1 are impaired in nef-expressing and HIV-1-infected astrocytes, suggesting that infection of astrocytes may play a significant role in the neuropathogenesis of HIV-1 encephalopathy.


Subject(s)
Astrocytes/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA Replication/drug effects , Endothelin-1/pharmacology , Gene Products, nef/metabolism , HIV-1/physiology , Astrocytoma , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , Cell Line , Enzyme Activation , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Gene Products, nef/biosynthesis , Humans , Mitogen-Activated Protein Kinase 1 , Protein-Tyrosine Kinases/metabolism , Receptors, CCR4 , Receptors, Chemokine/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Transfection , nef Gene Products, Human Immunodeficiency Virus
19.
Brain Res ; 768(1-2): 10-8, 1997 Sep 12.
Article in English | MEDLINE | ID: mdl-9369295

ABSTRACT

The immortalised RBE4 cell line, derived from rat brain capillary endothelial cells, preserves many features of the in vivo brain endothelium, and hence is of interest as a potential in vitro model of the blood-brain barrier (BBB). This study reports the effects of elevated intracellular cAMP and factors released by astrocytes on the F-actin cytoskeleton and paracellular sucrose permeability of monolayers of RBE4 cells. RBE4 cells grown in control medium showed a marked increase in the F-actin staining at the cytoplasmic margin at confluence, which was not significantly enhanced by elevation of intracellular cAMP and/or addition of astrocyte-conditioned medium (ACM). The formation of the marginal band of F-actin was accompanied by an increase in the F-actin content of the RBE4 cells up to confluence, and a decline in F-actin content thereafter. Elevation of intracellular cAMP or co-culture above astrocytes significantly decreased the paracellular sucrose permeability of confluent RBE4 cell monolayers grown on collagen filters (P < 0.01 and P < 0.001, respectively). Co-culture above astrocytes together with elevated cAMP also produced a significant decrease in the sucrose permeability of the monolayer (P < 0.01) but this was no greater than with astrocytes alone. These findings show that the RBE4 cell line may serve as a useful in vitro model for the study of brain endothelial cell physiology and agents which alter the permeability of the BBB.


Subject(s)
Actins/metabolism , Capillary Permeability/drug effects , Cyclic AMP/pharmacology , Cytoskeleton/drug effects , Endothelium, Vascular/drug effects , Sucrose/pharmacokinetics , Animals , Astrocytes/metabolism , Cell Line , Cell Survival/drug effects , Microcirculation/drug effects , Microscopy, Fluorescence , Rats
20.
J Neurosci ; 17(16): 6203-12, 1997 Aug 15.
Article in English | MEDLINE | ID: mdl-9236231

ABSTRACT

Endothelin-1 (ET-1) has been shown to induce DNA synthesis in primary astrocytes by stimulating the extracellular signal-regulated kinase (ERK) pathway. To clarify the mechanisms responsible for the anchorage-dependent growth of astrocytes, the relationships between cell adhesion and ERK activation were investigated. Here it is reported that ET-1 promotes the formation of stress fibers and focal adhesions and the tyrosine phosphorylation of focal adhesion kinase (FAK) and paxillin, as well as Src activation and association of phosphorylated FAK with Grb2. Pretreatment of astrocytes with cytochalasin D or C3-transferase, which inhibits actin polymerization or Rho activity, respectively, prevented the activation/phosphorylation of Src, FAK, and paxillin after ET-1 stimulation; by contrast, the ERK pathway was not significantly affected. This differential activation of FAK/Src and ERK pathways was also observed with astrocytes 10 and 60 min after replating on poly-L-ornithine-precoated dishes. Collectively, these findings indicate that activation of FAK and Src is dependent on actin cytoskeleton integrity, Rho activation, and adhesion to extracellular matrix, whereas ERK activation is independent of these intracellular events and seems to correlate with activation of the newly identified protein tyrosine kinase PYK2. Induction of DNA synthesis by ET-1, however, was reduced dramatically in astrocytes pretreated with either cytochalasin D or C3-transferase. This study provides a demonstration of Rho- and adhesion-dependent activation of FAK/Src, which collaborates with adhesion-independent activation of PYK2/ERK for DNA synthesis in ET-1-stimulated astrocytes.


Subject(s)
Adaptor Proteins, Signal Transducing , Astrocytes/cytology , Cell Adhesion Molecules/metabolism , Endothelin-1/pharmacology , Actins/metabolism , Aluminum Compounds/pharmacology , Animals , Astrocytes/chemistry , Astrocytes/enzymology , Cell Division/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Corpus Striatum/cytology , Cytoskeletal Proteins/metabolism , Cytoskeleton/physiology , DNA/biosynthesis , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Fetus/cytology , Fluorides/pharmacology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , GRB2 Adaptor Protein , GTP-Binding Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nerve Tissue Proteins/metabolism , Paxillin , Phosphoproteins/metabolism , Phosphorylation , Protein Binding/physiology , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins/chemistry , Proteins/metabolism , Rabbits , Rats , Signal Transduction/physiology , Stress, Mechanical , Tyrosine/metabolism , rho GTP-Binding Proteins , src Homology Domains/physiology , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...