Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
2.
Vet Microbiol ; 126(4): 383-9, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-17765409

ABSTRACT

The main objective of the present study was to investigate if different kinds of pig farms, like farrowing farms and rearing farms, play a role in the transmission of methicillin-resistant Staphylococcus aureus (MRSA) to Dutch finishing farms. Twelve farrowing farms, 11 finishing farms, 6 farrow-to finish farms, 1 rearing farm and 1 centre for artificial insemination were included. Screening of 310 pigs from these 31 farms showed 35 pigs (11%) to carry MRSA in their nares. On 7 of the 31 (23%) investigated farms colonized pigs were found, including 3 finishing farms, 3 farrowing farms and 1 farrow-to-finish farm. The use of standard antimicrobial medication of the pigs seemed to be a risk factor for MRSA carriage. Screening of the pigs on six farms supplying pigs for the MRSA positive farms revealed that the pigs on all but one farm were MRSA positive. Genotyping revealed that all MRSA strains were non-typeable by PFGE using the SmaI restriction enzyme and had multilocus sequence type (MLST) ST398. Different spa-types were found including t011, t108, t567, t899 and t1939, but the spa-types on epidemiologically related farms were identical indicating that MRSA are transmitted between farms through the purchase of colonized pigs. Two SCCmec types were found among the MRSA: type IV and type V. SCCmec type V was predominant. On two farms MRSA isolates with ST398, the same spa-type but with different SCCmec types (IV and V) were found, suggesting that different SCCmec elements have been inserted into MSSA with the same genotype. All MRSA strains were resistant to tetracycline, but additional resistances to erythromycin, lincomycin, kanamycin and gentamicin were also found. All MRSA isolates were negative for the exfoliative toxin genes (eta and etb), PVL toxin genes (lukF and lukS), toxic shock syndrome gene (tst-1), and the leukotoxin genes (lukE, lukD, lukM, lukF').


Subject(s)
Anti-Bacterial Agents/pharmacology , Methicillin Resistance , Staphylococcal Infections/veterinary , Staphylococcus aureus/drug effects , Swine Diseases/microbiology , Swine Diseases/transmission , Animals , Bacterial Typing Techniques/veterinary , Colony Count, Microbial/veterinary , Female , Genotype , Male , Methicillin/pharmacology , Microbial Sensitivity Tests/veterinary , Netherlands , Phylogeny , Prevalence , Staphylococcal Infections/microbiology , Staphylococcal Infections/transmission , Staphylococcus aureus/classification , Staphylococcus aureus/growth & development , Swine , Tandem Repeat Sequences
3.
J Neurovirol ; 13(1): 2-10, 2007.
Article in English | MEDLINE | ID: mdl-17454443

ABSTRACT

Although neurological symptoms associated with cerebral malaria (CM) are largely reversible, recent studies suggest that lasting neurological sequelae can occur. This may be especially true for children, in whom persistent deficits include problems with memory and attention. Because the malaria parasite is not thought to enter the brain parenchyma, lasting deficits are likely related to factors including the host response to disease. Studies with a rodent model, and with human postmortem tissue, suggest that glial activation occurs with CM. In this review, the authors will highlight studies focused on such activation in CM. Likely causes will be discussed, which include ischemia and activation of blood brain barrier endothelial cells. The potential consequences of glial activation will also be discussed, highlighting the possibility that glial-derived proteinases contribute to structural damage of the central nervous system (CNS). Of note, for the purposes of this focused review, glial activation will refer to the activation of astrocytes and microglial cells; discussion of oligodendroglial cells will not be included. In addition, although events thought to be critical to the pathogenesis of CM and glial activation will be covered, a comprehensive review of cerebral malaria will not be presented. Excellent reviews are already available, including Coltel et al (2004; Curr Neurovasc Res 1: 91-110), Medana and Turner (2006; Int J Parasitol 36: 555-568), and Hunt et al (2006; Int J Parasitol 36: 569-582).


Subject(s)
Central Nervous System Infections/cerebrospinal fluid , Central Nervous System/immunology , Malaria, Cerebral/enzymology , Malaria, Cerebral/pathology , Microglia/physiology , Animals , Central Nervous System/pathology , Central Nervous System Infections/physiopathology , Enzyme Activation , Humans , Malaria, Cerebral/immunology , Matrix Metalloproteinase 9/blood , Matrix Metalloproteinase 9/cerebrospinal fluid , Microglia/cytology
4.
Infect Immun ; 69(10): 6527-31, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11553600

ABSTRACT

One of the first steps in the development of cerebral toxoplasmosis is the penetration of the blood-brain barrier, which is comprised of microvascular endothelial cells. We examined the capacity of human brain microvascular endothelial cells (HBMEC) to interact with Toxoplasma gondii. We found that stimulation of HBMEC with gamma interferon (IFN-gamma) resulted in the induction of toxoplasmostasis. The capacity of HBMEC to restrict Toxoplasma growth after IFN-gamma stimulation was enhanced in the presence of tumor necrosis factor alpha (TNF-alpha). In addition, we found that IFN-gamma induced a strong induction of indoleamine 2,3-dioxygenase (IDO) activity in HBMEC, and this enzyme activity was enhanced by costimulation with TNF-alpha. The addition of excess amounts of tryptophan to the HBMEC cultures resulted in a complete abrogation of the IFN-gamma-TNF-alpha-mediated toxoplasmostasis. We therefore conclude that IDO induction contributed to the antiparasitic effector mechanism inducible in HBMEC by IFN-gamma and TNF-alpha.


Subject(s)
Brain/blood supply , Toxoplasma/growth & development , Tryptophan Oxygenase/metabolism , Adult , Animals , Capillaries/cytology , Cells, Cultured , Endothelium, Vascular/cytology , Enzyme Activation , Enzyme Induction , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Toxoplasma/drug effects , Tryptophan Oxygenase/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
5.
Neuropediatrics ; 32(4): 206-10, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11571701

ABSTRACT

Cerebral abscess is a rare complication of staphylococcal septicemia in infants associated with high mortality and morbidity. In the pathogenesis of abscess formation, S. aureus, one major causative agent, interacts with endothelial cells of the brain vessels before reaching the central nervous system. This study examined the growth of S. aureus in human brain microvascular endothelial cells (HBMEC) cultures stimulated with cytokines. IFN-gamma inhibited S. aureus replication by the induction of indoleamine 2,3-dioxygenase (IDO) in HBMEC. This activation of IDO in HBMEC could be shown by RT-PCR and by detection of kynurenine in culture supernatants of activated cells. Resupplementation of L-tryptophan abrogated the inhibitory effect of IFN-gamma on the growth of staphylococci, hence confirming the activation of indoleamine 2,3-dioxygenase as being responsible for the induced bacteriostasis. Addition of TNF-alpha enhanced the IFN-gamma mediated antibacterial effects, whereas TNF-alpha alone had no influence on staphylococcal growth. Stimulation of HBMEC with IFN-gamma failed to activate inducible nitric oxide synthase (iNOS) and subsequent production of nitric oxide (NO). Thus, intra- and extracellular depletion of L-tryptophan seems to be an important process in the defense against staphylococcal brain abscesses by means of creating an unfavorable microenvironment.


Subject(s)
Brain Abscess/immunology , Endothelium, Vascular/immunology , Interferon-gamma/pharmacology , Staphylococcus aureus/drug effects , Tryptophan Oxygenase/metabolism , Tryptophan/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Brain Abscess/microbiology , Cell Culture Techniques , Cytokines/pharmacology , Drug Therapy, Combination , Endothelium, Vascular/enzymology , Endothelium, Vascular/microbiology , Enzyme Activation/drug effects , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Reverse Transcriptase Polymerase Chain Reaction , Staphylococcus aureus/immunology , Tryptophan Oxygenase/biosynthesis
6.
J Neurovirol ; 7(2): 125-34, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11517385

ABSTRACT

Encephalopathy represents a common and serious manifestation of HIV-1 infection in children, but its pathogenesis is unclear. We demonstrated that gp120 activated human brain microvascular endothelial cells (HBMEC) derived from children in up-regulating ICAM-1 and VCAM-1 expression, IL-6 secretion and increased monocyte transmigration across monolayers. Another novel observation was our demonstration of CD4 in isolated HBMEC and on microvessels of children's brain cryosections. Gp120-induced monocyte migration was inhibited by anti-gp120 and anti-CD4 antibodies. This is the first demonstration that gp120 activates HBMEC via CD4, which may contribute to the development of HIV-1 encephalopathy in children.


Subject(s)
AIDS Dementia Complex/virology , Brain/virology , CD4 Antigens/metabolism , Endothelium, Vascular/virology , HIV Envelope Protein gp120/metabolism , HIV-1 , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/pathology , Adult , Antibodies/pharmacology , Brain/blood supply , Brain/pathology , CD4 Antigens/genetics , CD4 Antigens/immunology , Cell Movement/immunology , Child , Child, Preschool , Endothelium, Vascular/chemistry , Endothelium, Vascular/metabolism , Flow Cytometry , Frozen Sections , HIV Envelope Protein gp120/immunology , Humans , In Vitro Techniques , Intercellular Adhesion Molecule-1/analysis , Intercellular Adhesion Molecule-1/biosynthesis , Monocytes/cytology , Monocytes/immunology , Monocytes/virology , RNA, Messenger/analysis , Vascular Cell Adhesion Molecule-1/analysis , Vascular Cell Adhesion Molecule-1/biosynthesis
7.
J Infect Dis ; 184(6): 732-7, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11517434

ABSTRACT

Invasion of brain microvascular endothelial cells (BMECs) is a key step in the pathogenesis of meningitis due to Escherichia coli and Listeria monocytogenes. Although host cell actin cytoskeletal rearrangements are essential in BMEC invasion by E. coli K1 and L. monocytogenes, the underlying signaling mechanisms remain unclear. This study demonstrates that host cell cytosolic phospholipase A2 (cPLA2) contributes to E. coli K1 invasion of BMECs but not to L. monocytogenes invasion of BMECs. This difference was observed with 4-bromophenacyl bromide, a nonselective PLA2 inhibitor, and arachidonyl trifluoromethyl ketone, a selective cPLA2 inhibitor, and was confirmed with BMEC derived from cPLA2 knockout mice. Activation of cPLA2 leads to generation of intracellular arachidonic acid, which is metabolized via cyclooxygenase (COX) and lipo-oxygenase (LOX) pathways into eicosanoids. COX and LOX inhibitors also significantly inhibit E. coli K1 invasion of BMECs.


Subject(s)
Benzylisoquinolines , Cerebrovascular Circulation/physiology , Endothelium, Vascular/enzymology , Endothelium, Vascular/microbiology , Escherichia coli/pathogenicity , Listeria monocytogenes/pathogenicity , Phospholipases A/metabolism , Alkaloids/pharmacology , Arachidonic Acids/pharmacology , Cells, Cultured , Cerebral Cortex/blood supply , Child , Child, Preschool , Cytosol/enzymology , Drugs, Chinese Herbal/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , Escherichia coli/physiology , Humans , Indomethacin/pharmacology , Lipoxygenase Inhibitors/pharmacology , Listeria monocytogenes/drug effects , Listeria monocytogenes/physiology , Masoprocol/pharmacology , Microcirculation/enzymology , Microcirculation/microbiology , Phospholipases A/antagonists & inhibitors , Phospholipases A2 , Signal Transduction
8.
Cancer Res ; 61(14): 5587-94, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454712

ABSTRACT

Increased expression of plasminogen activator inhibitor-1 (PAI-1) in cancer patients is associated with unfavorable outcome, and the reason for this paradox has been poorly understood. We have previously reported elevated levels of PAI-1 in primary tumors of advanced neuroblastomas (Y. Sugiura et al., Cancer Res., 59: 1327-1336, 1999). Here we demonstrate that PAI-1 is coexpressed with the angiogenesis marker alpha(v)beta3 integrin in blood vessels of primary neuroblastoma tumors, suggesting that PAI-1 plays a role in angiogenesis. Using human brain microvascular endothelial cells (HBMECs), we found that PAI-1 inhibits alpha(v)beta3 integrin-mediated cell adhesion to vitronectin but promotes alpha5beta1-mediated migration from vitronectin toward fibronectin. Inhibition of vitronectin adhesion by PAI-1 did not induce HBMEC apoptosis. PAI-1 also inhibited endothelial tube formation on Matrigel in the presence of vitronectin but had a stimulatory effect in the presence of fibronectin. This effect of PAI-1 on microvascular endothelial cells is primarily related to the ability of PAI-1 to bind to vitronectin via its NH2-terminal domain and to interfere with cell adhesion to vitronectin. We propose that PAI-1 acts as a positive switch for angiogenesis by promoting endothelial cell migration away from their vitronectin-containing perivascular space toward fibronectin-rich tumor tissue. These observations provide a novel explanation for the enhancing effect of PAI-1 in cancer progression.


Subject(s)
Cell Movement/drug effects , Endothelium, Vascular/drug effects , Fibronectins/pharmacology , Neovascularization, Pathologic/pathology , Plasminogen Activator Inhibitor 1/pharmacology , Apoptosis/drug effects , Cell Adhesion/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Humans , Immunohistochemistry , Integrins/physiology , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/drug effects , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/physiology , Recombinant Proteins/pharmacology , Vitronectin/pharmacology
9.
Infect Immun ; 69(7): 4536-44, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11401997

ABSTRACT

Candida albicans is an opportunistic pathogen, which primarily affects neonates and immunocompromised individuals. The pathogen can invade the central nervous system, resulting in meningitis. At present, the pathogenesis of C. albicans meningitis is unclear. We used an in vitro model of the human blood-brain barrier to investigate the interaction(s) of C. albicans with human brain microvascular endothelial cells (BMEC). Binding of C. albicans to human BMEC was time and inoculum dependent. Invasion of C. albicans into human BMEC was demonstrated by using an enzyme-linked immunosorbent assay based on fluorescent staining of C. albicans with calcoflour. In contrast, avirulent Candida mutant strains and nonpathogenic yeast Saccharomyces cerevisiae were not able to bind and invade human BMEC. Morphological studies revealed that on association with human BMEC, C. albicans formed germ tubes and was able to bud intracellularly. Transmission electron microscopy showed various stages of C. albicans interactions with human BMEC, e.g., pseudopod-like structures on human BMEC membrane and intracellular vacuole-like structures retaining C. albicans. Of interest, C. albicans was able to bud and develop pseudohyphae inside human BMEC without apparent morphological changes of the host cells. In addition, C. albicans penetrates through human BMEC monolayers without a detectable change in transendothelial electrical resistance and inulin permeability. This is the first demonstration that C. albicans is able to adhere, invade, and transcytose across human BMEC without affecting monolayer integrity. A complete understanding of the interaction(s) of C. albicans with human BMEC should contribute to the understanding of the pathogenic mechanism(s) of C. albicans meningitis.


Subject(s)
Blood-Brain Barrier , Candida albicans/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Microscopy, Electron
10.
Clin Immunol ; 99(3): 387-94, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11358436

ABSTRACT

Leukocyte endothelial adhesion (LEA) is the prelude to a complex cascade of reactions following an immunological challenge. Recently, LEA has been implicated in the molecular basis of several dermatological disorders. While the role of proinflammatory cytokines, such as interleukin-1 beta (IL-1beta) and tumor necrosis factor-alpha (TNF-alpha), in LEA has been investigated using nondermal models, limited data exist regarding their effects on LEA in dermal models. This study shows that cotreatment of cultured human dermal endothelial cells (CADMEC) with IL-1beta and TNF-alpha resulted in a marked increase in the adherence of human promyelocytic leukemia (HL-60) cells to CADMEC and an increase in expression of intercellular adhesion molecule-1 and E-selectin. Pretreatment of CADMEC with dexamethasone, a long-lasting glucocorticoid, resulted in a decrease in both HL-60 cell adhesion to CADMEC and adhesion molecule expression. Taken together, these data demonstrate that LEA may play a role in inflammatory skin conditions and in the mechanisms underlying the potential use of glucocorticoids as a treatment option.


Subject(s)
Dexamethasone/pharmacology , Endothelium, Vascular/cytology , Interleukin-1/pharmacology , Skin/immunology , Tumor Necrosis Factor-alpha/pharmacology , Cell Adhesion/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , E-Selectin/analysis , HL-60 Cells , Humans , Intercellular Adhesion Molecule-1/analysis , Leukocytes/physiology , Skin/blood supply , Vascular Cell Adhesion Molecule-1/analysis
11.
Microb Pathog ; 30(1): 19-28, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11162182

ABSTRACT

Most cases of neonatal bacterial meningitis develop as a result of a hematogenous spread, but it is not clear how circulating bacteria cross the blood-brain barrier. Attempts to answer these questions have been hampered by the lack of a reliable model of the human blood-brain barrier. Human brain microvascular endothelial cells (HBMEC) were isolated and transfected with a pBR322 based plasmid containing simian virus 40 large T antigen (SV40-LT). The transfected HBMEC exhibited similar brain endothelial cell characteristics as the primary HBMEC, i.e. gamma glutamyl transpeptidase and a high transendothelial electrical resistance. Escherischia coli and Citrobacter spp, two important Gram-negative bacilli causing neonatal meningitis, were found to transcytose across primary and transfected HBMEC, without affecting the integrity of the monolayer. In addition, E. coli and C. freundii invaded transfected HBMEC as shown previously with primary HBMEC. We conclude that E. coli and C. freundii are able to invade and transcytose HBMEC and these bacterial-HBMEC interactions are similar between primary and transfected HBMEC. Therefore, our transfected HBMEC should be useful for studying pathogenesis of CNS infections.


Subject(s)
Brain/blood supply , Citrobacter freundii/physiology , Citrobacter freundii/pathogenicity , Endothelium, Vascular/microbiology , Escherichia coli/physiology , Escherichia coli/pathogenicity , Blood-Brain Barrier , Cells, Cultured , Endothelium, Vascular/cytology , Enterobacteriaceae Infections/microbiology , Escherichia coli Infections/microbiology , Humans , Microcirculation , Transfection
12.
Am J Physiol Cell Physiol ; 279(6): C1772-81, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11078691

ABSTRACT

In patients with amyloid beta-related cerebrovascular disorders, e.g. , Alzheimer's disease, one finds increased deposition of amyloid peptide (Abeta) and increased presence of monocyte/microglia cells in the brain. However, relatively little is known of the role of Abeta in the trafficking of monocytes across the blood-brain barrier (BBB). Our studies show that interaction of Abeta(1-40) with monolayer of human brain endothelial cells results in augmented adhesion and transendothelial migration of monocytic cells (THP-1 and HL-60) and peripheral blood monocytes. The Abeta-mediated migration of monocytes was inhibited by antibody to Abeta receptor (RAGE) and platelet endothelial cell adhesion molecule (PECAM-1). Additionally, Abeta-induced transendothelial migration of monocytes were inhibited by protein kinase C inhibitor and augmented by phosphatase inhibitor. We conclude that interaction of Abeta with RAGE expressed on brain endothelial cells initiates cellular signaling leading to the transendothelial migration of monocytes. We suggest that increased diapedesis of monocytes across the BBB in response to Abeta present either in the peripheral circulation or in the brain parenchyma may play a role in the pathophysiology of Abeta-related vascular disorder.


Subject(s)
Amyloid beta-Peptides/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Monocytes/cytology , Peptide Fragments/pharmacology , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Receptors, Immunologic/immunology , Adult , Anti-Inflammatory Agents/pharmacology , Antibodies/pharmacology , Anticoagulants/pharmacokinetics , Blood-Brain Barrier/immunology , Brain/blood supply , Brain/metabolism , Capillaries/cytology , Capillaries/metabolism , Carbon Radioisotopes , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Movement/drug effects , Cell Movement/immunology , Cell Survival/drug effects , Cells, Cultured , Dextrans/pharmacokinetics , Endothelium, Vascular/immunology , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , HL-60 Cells/cytology , Humans , Indoles/pharmacology , Inulin/pharmacokinetics , Maleimides/pharmacology , Receptor for Advanced Glycation End Products
13.
Microbes Infect ; 2(10): 1237-44, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11008113

ABSTRACT

Bacterial pathogens may breach the blood-brain barrier (BBB) and invade the central nervous system through paracellular and/or transcellular mechanisms. Transcellular penetration, e.g., transcytosis across the BBB has been demonstrated for Escherichia coli K1, group B streptococcus, Listeria monocytogenes, Citrobacter freundii and Streptococcus pneumonia strains. Genes contributing to invasion of brain microvascular endothelial cells include E. coli K1 genes ompA, ibeA, ibeB, and yijP. Understanding the mechanisms of bacterial penetration across the BBB may help develop novel approaches to preventing bacterial meningitis.


Subject(s)
Blood-Brain Barrier , Brain/microbiology , Meningitis, Bacterial/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Brain/pathology , Brain/ultrastructure , Citrobacter/genetics , Citrobacter/pathogenicity , Endothelium, Vascular/microbiology , Endothelium, Vascular/pathology , Escherichia coli/genetics , Escherichia coli/pathogenicity , Humans , Infant, Newborn , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Meningitis, Bacterial/pathology , Streptococcus/genetics , Streptococcus/pathogenicity
14.
Cancer Res ; 60(3): 712-21, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10676658

ABSTRACT

Inhibition of the RGD-binding integrins, alpha(v)beta3 and alpha(v)beta5, prevents endothelial cell anchorage and induces endothelial apoptosis, which results in disruption of tumor angiogenesis and inhibition of tumor growth in animal models. In this study, we demonstrate by immunohistochemical analysis that integrin alpha(v)beta3 was expressed by 61% (mean) of microvessels in high-risk neuroblastomas (stage IV and MYCN-amplified stage III; n = 28) but only by 18% (mean) of microvessels in low-risk tumors (stages I and II and non-MYCN-amplified stage III; n = 12). Integrin alpha(v)beta5 was found on 60% (mean) of microvessels in 21 Stage IV tumors. These data suggest that neuroblastomas may be targeted for antiangiogenic treatment directed against endothelial integrins alpha(v)beta3 and alpha(v)beta5. In cell culture, inhibition of integrin-dependent endothelial cell anchorage to vitronectin by RGDfV, an RGD function-blocking cyclic peptide, induced apoptosis in bovine brain endothelial cells compared with the control peptide, RADfV (37.5% versus 8.7%, respectively), as detected by chromatin condensation and nuclear fragmentation. Treatment with RGDfV but not with RADfV, which prevented attachment of endothelial cells to vitronectin or fibronectin, was associated with up to a 50% increase in endogenous ceramide, a lipid second messenger that can mediate cell death. Furthermore, exogenous C2-ceramide was cytotoxic to bovine brain endothelial cells and induced activation of C-jun N-terminal kinase (JNK), a MAP kinase that can be activated in stress-induced apoptosis pathways. This suggests that ceramide may function in detachment-induced endothelial cell apoptosis, originating from inhibition of vitronectin binding to integrins such as alpha(v)beta3 and alpha(v)beta5. This is the first report to demonstrate expression of integrins alpha(v)beta3 and alpha(v)beta5 by microvascular endothelium of a childhood tumor and association of their expression with neuroblastoma aggressiveness. Furthermore, our data provide the first suggestion that inhibition of endothelial cell anchorage, resulting from specific blockade of RGD-binding integrins, increases endogenous ceramide, which may contribute to endothelial cell death.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Ceramides/biosynthesis , Endothelium, Vascular/metabolism , Integrins/physiology , JNK Mitogen-Activated Protein Kinases , Neuroblastoma/metabolism , Receptors, Vitronectin/physiology , Apoptosis , Endothelium, Vascular/cytology , Enzyme Activation , Humans , Immunohistochemistry , Integrins/analysis , Integrins/antagonists & inhibitors , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases/metabolism , Neuroblastoma/drug therapy , Receptors, Vitronectin/analysis , Receptors, Vitronectin/antagonists & inhibitors
15.
Am J Pathol ; 155(5): 1599-611, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10550317

ABSTRACT

The numbers of immune-activated brain mononuclear phagocytes (MPs) affect the progression of human immunodeficiency virus (HIV)-1-associated dementia (HAD). Such MPs originate, in measure, from a pool of circulating monocytes. To address the mechanism(s) for monocyte penetration across the blood-brain barrier (BBB), we performed cross-validating laboratory, animal model, and human brain tissue investigations into HAD pathogenesis. First, an artificial BBB was constructed in which human brain microvascular endothelial and glial cells-astrocytes, microglia, and/or monocyte-derived macrophages (MDM)-were placed on opposite sides of a matrix-coated porous membrane. Second, a SCID mouse model of HIV-1 encephalitis (HIVE) was used to determine in vivo monocyte blood-to-brain migration. Third, immunohistochemical analyses of human HIVE tissue defined the relationships between astrogliosis, activation of microglia, virus infection, monocyte brain infiltration, and beta-chemokine expression. The results, taken together, showed that HIV-1-infected microglia increased monocyte migration through an artificial BBB 2 to 3.5 times more than replicate numbers of MDM. In the HIVE SCID mice, a marked accumulation of murine MDM was found in areas surrounding virus-infected human microglia but not MDM. For human HIVE, microglial activation and virus infection correlated with astrogliosis, monocyte transendothelial migration, and beta-chemokine expression. Pure cultures of virus-infected and activated microglia or astrocytes exposed to microglial conditioned media produced significant quantities of beta-chemokines. We conclude that microglial activation alone and/or through its interactions with astrocytes induces beta-chemokine-mediated monocyte migration in HAD.


Subject(s)
AIDS Dementia Complex/pathology , Blood-Brain Barrier , Cell Movement , Chemokines/metabolism , HIV-1 , Monocytes/pathology , AIDS Dementia Complex/metabolism , Adult , Aged , Animals , Astrocytes/metabolism , Astrocytes/pathology , Child , Child, Preschool , Humans , Immunohistochemistry , Male , Mice , Mice, SCID , Microglia/metabolism , Microglia/pathology , Middle Aged , Monocytes/metabolism
16.
Neurosurgery ; 45(4): 962-8; discussion 968-70, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10515502

ABSTRACT

OBJECTIVES: Gene transfer of thrombolytic enzymes to vascular endothelial cells may influence the kinetics of intravascular thrombosis. This study defines the potential for gene transfer of tissue plasminogen activator (tPA) into bovine brain endothelial cells (BBEC). METHODS: The retroviral vectors derived from murine leukemia virus (MuLV) were used to transfer human tPA cDNA to BBEC. The tPA activity, tPA antigen and tPA inhibitor 1 (PAI-1) antigen were determined in the supernatant of transduced (BBEC/tPA) cell cultures by an immunoassay. RESULTS: The tPA antigen and enzymatic activity in cell culture supernatants of BBEC/tPA transduced cells were 75 ng/ml and 14 IU/ml after 4 days, that was 25 and 28-fold higher compared to the respective values in control cells. The PAI-1 antigen was not affected by tPA cDNA transfer. The Western blot assay of cell lysates confirmed that the majority of tPA in BBEC/tPA transduced cells was in the form of free tPA. While the maximal transduction efficiency of BBEC with an amphotropic MuLV vector was about 15%, a MuLV pseudotyped with vesicular stomatitis virus G glycoprotein envelope achieved high > 90% maximal transduction efficiency. CONCLUSIONS: The fibrinolytic activity of brain endothelial cells can be enhanced by transferring human tPA cDNA. These findings provide an initial step in implementation of future studies that investigate the use of this technology as an adjunctive treatment for cerebrovascular disease.


Subject(s)
Brain/blood supply , DNA, Complementary/genetics , Endothelium, Vascular/metabolism , Gene Transfer Techniques , Tissue Plasminogen Activator/genetics , Animals , Cattle , Cells, Cultured , Fibrinolysis/genetics , Fibrinolysis/physiology , Humans , Retroviridae/genetics
17.
Infect Immun ; 67(11): 5775-83, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10531228

ABSTRACT

Escherichia coli is the most common gram-negative bacterium that causes meningitis during the neonatal period. We have previously shown that the entry of circulating E. coli organisms into the central nervous system is due to their ability to invade the blood-brain barrier, which is composed of a layer of brain microvascular endothelial cells (BMEC). In this report, we show by transmission electron microscopy that E. coli transmigrates through BMEC in an enclosed vacuole without intracellular multiplication. The microfilament-disrupting agents cytochalasin D and latrunculin A completely blocked E. coli invasion of BMEC. Cells treated with the microtubule inhibitors nocodazole, colchicine, vincristin, and vinblastine and the microtubule-stabilizing agent taxol also exhibited 50 to 60% inhibition of E. coli invasion. Confocal laser scanning fluorescence microscopy showed F-actin condensation associated with the invasive E. coli but no alterations in microtubule distribution. These results suggest that E. coli uses a microfilament-dependent phagocytosis-like endocytic mechanism for invasion of BMEC. Previously we showed that OmpA expression significantly enhances the E. coli invasion of BMEC. We therefore examined whether OmpA expression is related to the recruitment of F-actin. OmpA(+) E. coli induced the accumulation of actin in BMEC to a level similar to that induced by the parental strain, whereas OmpA(-) E. coli did not. Despite the presence of OmpA, a noninvasive E. coli isolate, however, did not show F-actin condensation. OmpA(+)-E. coli-associated condensation of F-actin was blocked by synthetic peptides corresponding to the N-terminal extracellular domains of OmpA as well as BMEC receptor analogues for OmpA, chitooligomers (GlcNAcbeta1-4GlcNAc oligomers). These findings suggest that OmpA interaction is critical for the expression or modulation of other bacterial proteins that will subsequently cause actin accumulation for the uptake of bacteria.


Subject(s)
Actins/metabolism , Bacterial Outer Membrane Proteins/physiology , Brain/microbiology , Endothelium, Vascular/microbiology , Escherichia coli/physiology , Animals , Brain/blood supply , Cattle , Clathrin/physiology , Cytochalasin D/pharmacology , Endocytosis , Endothelium, Vascular/ultrastructure , Microscopy, Electron , Microtubules/physiology
18.
Pharm Res ; 16(9): 1360-5, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10496650

ABSTRACT

PURPOSE: To study Cereport (RMP-7, bradykinin B2 agonist) effects on human brain microvascular endothelial cell (HBMEC) monolayer permeability. METHODS: HBMEC grown on transwell membranes were exposed to Cereport. The monolayer permeability was determined with [14C]-inulin (MW. 5,200) and [3H]-dextran (MW. 70,000). RESULTS: Cereport increased the HBMEC permeability to [14C]-inulin, but not to [3H]-dextran. The effect was transient, maximal at 15 min (i.e., 79.3% increase), and polarized to the basolateral membrane. An inverted U, dose-response curve was observed with active concentrations of Cereport from 0.01 to 0.5 nmol/L, the plateau maximal effect between 0.5 and 10 nmol/L, and loss of activity at the highest concentration, i.e., 20 nmol/L. Cyclic AMP-specific phosphodiesterase 3 (PDE3) inhibitor rolipram (10 micromol/L) abolished Cereport effects, while cGMP-specific PDE5 inhibitor, zaniprast (50 micromol/L) enhanced by 31% (p < 0.05) the effect of 0.1 nmolL Cereport. Unlabeled Cereport displaced [125I]-bradykinin and/or [125I]-Cereport from the basolateral side. There was no specific Cereport binding to the apical side. CONCLUSIONS: Cereport exerts specific time, dose and size dependent actions on HMBEC monolayer that are restricted to the basolateral membrane. Its effects can be further modulated through changes in cAMP and cGMP second messenger systems.


Subject(s)
Blood-Brain Barrier/drug effects , Bradykinin/analogs & derivatives , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Inulin/pharmacokinetics , Binding, Competitive , Bradykinin/metabolism , Bradykinin/pharmacology , Carbon Radioisotopes , Cells, Cultured , Cerebral Cortex/blood supply , Dextrans/pharmacokinetics , Humans , Iodine Radioisotopes , Microcirculation/drug effects , Phosphodiesterase Inhibitors/pharmacology , Purinones/pharmacology , Receptor, Bradykinin B2 , Receptors, Bradykinin/physiology , Rolipram/pharmacology , Tritium
19.
Infect Immun ; 67(10): 5522-5, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10496943

ABSTRACT

Escherichia coli meningitis commonly occurs in the neonatal period, but the basis of this age dependency is unclear. We have previously identified two types of E. coli-brain microvascular endothelial cell (BMEC) interactions contributing to E. coli traversal of the blood-brain barrier (i.e., binding and invasion). The present study examined whether the age dependency of E. coli meningitis stemmed from differences in the capacities of neonatal and adult BMECs to interact with E. coli. BMECs were isolated from rats of different ages (10 days, 20 days and 3 months) as well as from humans of different ages (fetuses, 4- to 7-year-old children, and a 35-year-old adult, and 60- to 85-year-old geriatrics). The bindings of E. coli to young and old rat BMECs were similar. Also, the abilities of E. coli to invade BMECs were similar for BMECs derived from young and old rats and from human fetuses, children, adults, and geriatrics. These findings suggest that the predominance of E. coli meningitis in neonates is not likely due to greater binding and invasion capacities of newborn compared to adult BMECs.


Subject(s)
Bacterial Adhesion , Brain/blood supply , Brain/microbiology , Endothelium, Vascular/microbiology , Escherichia coli/physiology , Meningitis, Bacterial/etiology , Adult , Age Factors , Aged , Animals , Child , Humans , Rats
20.
Infect Immun ; 67(9): 4751-6, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10456927

ABSTRACT

Most cases of Escherichia coli meningitis develop as a result of hematogenous spread, but it is not clear how circulating E. coli crosses the blood-brain barrier. A TnphoA mutant of E. coli K1 RS218 was shown to be significantly less invasive than its parent strain in bovine and human brain microvascular endothelial cells (BMEC), which constitute the blood-brain barrier. More importantly, traversal of the blood-brain barrier was significantly less with this mutant than with the parent strain in newborn rats with experimental hematogenous meningitis. A DNA segment containing the TnphoA insertion site was cloned from RS218, and the cloned DNA complemented the TnphoA mutant in invasion of BMEC. Nucleotide sequence revealed a near identity to that of a hypothetical yijP gene (also called f577) in the E. coli K-12 genome. Sequence analysis indicated that the E. coli K1 yijP gene likely encodes a 66. 6-kDa membrane protein. Deletion and complementation experiments indicated that the yijP gene was involved in E. coli K1 invasion of BMEC, i.e., the invasive ability of E. coli K1 was significantly reduced after yijP was deleted and was restored by complementation with a plasmid containing the yijP open reading frame. This is the first demonstration that the yijP gene locus plays a role in the pathogenesis of E. coli K1 meningitis.


Subject(s)
Bacterial Proteins/genetics , Brain/microbiology , Endothelium, Vascular/microbiology , Escherichia coli Infections/microbiology , Escherichia coli Proteins , Escherichia coli/genetics , Membrane Proteins , Meningitis, Bacterial/microbiology , Animals , Bacterial Proteins/physiology , Base Sequence , Blood-Brain Barrier , Brain/blood supply , Cattle , Chromosome Deletion , Chromosome Mapping , Chromosomes, Bacterial , Cloning, Molecular , DNA, Bacterial , Endothelium, Vascular/cytology , Escherichia coli/isolation & purification , Escherichia coli/pathogenicity , Genes, Bacterial , Genetic Complementation Test , Humans , Molecular Sequence Data , Mutagenesis, Insertional , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...