Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Commun ; 10(1): 1997, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31040288

ABSTRACT

Human G protein-coupled receptors (GPCRs) respond to various ligands and stimuli. However, GPCRs rely on membrane for proper folding, making their biochemical properties difficult to study. By displaying GPCRs in viral envelopes, we fabricated a Virion Display (VirD) array containing 315 non-olfactory human GPCRs for functional characterization. Using this array, we found that 10 of 20 anti-GPCR mAbs were ultra-specific. We further demonstrated that those failed in the mAb assays could recognize their canonical ligands, suggesting proper folding. Next, using two peptide ligands on the VirD-GPCR array, we identified expected interactions and novel interactions. Finally, we screened the array with group B Streptococcus, a major cause of neonatal meningitis, and demonstrated that inhibition of a newly identified target, CysLTR1, reduced bacterial penetration both in vitro and in vivo. We believe that the VirD-GPCR array holds great potential for high-throughput screening for small molecule drugs, affinity reagents, and ligand deorphanization.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Virion/metabolism , Animals , Blotting, Western , Chlorocebus aethiops , Fluorescent Antibody Technique , HEK293 Cells , HeLa Cells , Humans , Proteomics/methods , Streptococcus/metabolism , Vero Cells , Virology/methods
2.
Anal Chem ; 90(18): 10958-10966, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30106562

ABSTRACT

Bacterial meningitis in neonates and infants is an acute lethal disease and occurs in response to microbial exploitation of the blood-brain barrier (BBB), resulting in the intracranial inflammation. Several pathogens, such as Escherichia coli ( E. coli), can cause this devastating disease; however, the underlying molecular mechanisms by which these pathogens exploit the BBB remain incompletely understood. To identify important players on both the pathogen and host sides that govern the E. coli-BBB cell interactions, we took advantage of the E. coli and human proteome microarrays (i.e., HuProt) as an unbiased, proteome-wide tool for identification of important players on both sides. Using the E. coli proteome microarrays, we developed a unique high throughput chip-based cell probing assay to probe with fluorescent live human brain microvascular endothelial cells (HBMEC, which constitute the BBB). We identified several transmembrane proteins, which effectively bound to live HBMEC. We focused on YojI protein for further study. By probing the HuProt arrays with YojI, interferon-alpha receptor (IFNAR2) was identified as one of its binding proteins. The importance of YojI and IFNAR2 involved in E. coli-HBMEC interactions was characterized using the YojI knockout bacteria and IFNAR2-knock down HBMEC and further confirmed by E. coli binding assay in HBMEC. This study represents a new paradigm (dual-microarray technology) that enables rapid, unbiased discovery of both pathogen and host players that are involved in pathogen-host interactions for human infectious diseases in a high throughput manner.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Blood-Brain Barrier/microbiology , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Host-Pathogen Interactions , Proteomics/instrumentation , Receptor, Interferon alpha-beta/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Cell Line , Equipment Design , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Humans , Lab-On-A-Chip Devices
3.
Cell Microbiol ; 14(10): 1544-53, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22646320

ABSTRACT

Cryptococcus neoformans penetration into the central nervous system (CNS) requires traversal of the blood-brain barrier that is composed of a single layer of human brain microvascular endothelial cells (HBMEC), but the underlying mechanisms of C. neoformans traversal remain incompletely understood. C. neoformans transcytosis of HBMEC monolayer involves rearrangements of the host cell actin cytoskeleton and small GTP-binding Rho family proteins such as Rac1 are shown to regulate host cell actin cytoskeleton. We, therefore, examined whether C. neoformans traversal of the blood-brain barrier involves host Rac1. While the levels of activated Rac1 (GTP-Rac1) in HBMEC increased significantly upon incubation with C. neoformans strains, pharmacological inhibition and down-modulation of Rac1 significantly decreased C. neoformans transcytosis of HBMEC monolayer. Also, Rac1 inhibition was efficient in preventing C. neoformans penetration into the brain. In addition, C. neoformans phospholipase B1 (Plb1) was shown to contribute to activating host cell Rac1, andSTAT3 was observed to associate with GTP-Rac1 in HBMEC that were incubated with C. neoformans strain but not with its Δplb1 mutant. These findings demonstrate for the first time that C. neoformans Plb1 aids fungal traversal across the blood-brain barrier by activating host cell Rac1 and its association with STAT3, and suggest that pharmacological intervention of host-microbial interaction contributing to traversal of the blood-brain barrier may prevent C. neoformans penetration into the brain.


Subject(s)
Blood-Brain Barrier/microbiology , Cryptococcus neoformans/pathogenicity , Host-Pathogen Interactions , Phospholipases/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Line , Endothelial Cells/microbiology , Endothelial Cells/physiology , Gene Expression Profiling , Humans , Protein Binding , STAT3 Transcription Factor/metabolism , Transcytosis
4.
Infect Immun ; 79(10): 4088-93, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21825068

ABSTRACT

Group B Streptococcus (GBS) is the most common bacterium causing neonatal meningitis, and neonatal GBS meningitis continues to be an important cause of mortality and morbidity. Here we provide the first direct evidence that host cytosolic phospholipase A2α (cPLA2α) contributes to type III GBS invasion of human brain microvascular endothelial cells (HBMEC), which constitute the blood-brain barrier and penetration into the brain, the key step required for the development of GBS meningitis. This was shown by our demonstration that pharmacological inhibition and gene deletion of cPLA2α significantly decreased GBS invasion of the HBMEC monolayer and penetration into the brain. cPLA2α releases arachidonic acid from membrane phospholipids, and we showed that the contribution of cPLA2α to GBS invasion of HBMEC involved lipoxygenated metabolites of arachidonic acid, cysteinyl leukotrienes (LTs). In addition, type III GBS invasion of the HBMEC monolayer involves protein kinase Cα (PKCα), as shown by time-dependent PKCα activation in response to GBS as well as decreased GBS invasion in HBMEC expressing dominant-negative PKCα. PKCα activation in response to GBS, however, was abolished by inhibition of cPLA2α and cysteinyl LTs, suggesting that cPLA2α and cysteinyl LTs contribute to type III GBS invasion of the HBMEC monolayer via PKCα. These findings demonstrate that specific host factors involving cPLA2α and cysteinyl LTs contribute to type III GBS penetration of the blood-brain barrier and their contribution involves PKCα.


Subject(s)
Blood-Brain Barrier/microbiology , Cysteine/metabolism , Endothelial Cells/enzymology , Leukotrienes/metabolism , Phospholipases A2, Cytosolic/metabolism , Protein Kinase C-alpha/metabolism , Streptococcus agalactiae/pathogenicity , Arachidonic Acid/metabolism , Brain/blood supply , Brain/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/microbiology , Enzyme Activation , Humans , Microcirculation , Phospholipases A2, Cytosolic/genetics , Protein Kinase C-alpha/genetics , Streptococcus agalactiae/metabolism
5.
Infect Immun ; 78(8): 3554-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20515928

ABSTRACT

Escherichia coli meningitis is an important cause of mortality and morbidity, and a key contributing factor is our incomplete understanding of the pathogenesis of E. coli meningitis. We have shown that E. coli penetration into the brain requires E. coli invasion of human brain microvascular endothelial cells (HBMEC), which constitute the blood-brain barrier. E. coli invasion of HBMEC involves its interaction with HBMEC receptors, such as E. coli cytotoxic necrotizing factor 1 (CNF1) interaction with its receptor, the 67-kDa laminin receptor (67LR), and host signaling molecules including cytosolic phospholipase A(2)alpha (cPLA(2)alpha). In the present study, we showed that treatment with etoposide resulted in decreased expression of 67LR on HBMEC and inhibited E. coli invasion of HBMEC. Pharmacological inhibition of cysteinyl leukotrienes, lipoxygenated products of arachidonic acid released by cPLA(2)alpha, using montelukast (an antagonist of the type 1 cysteinyl leukotriene receptor) also inhibited E. coli invasion of HBMEC. E. coli penetration into the brain was significantly decreased by etoposide as well as by montelukast, and a combination of etoposide and montelukast was significantly more effective in inhibiting E. coli K1 invasion of HBMEC than single agents alone. These findings demonstrate for the first time that counteracting the HBMEC receptor and signaling molecule involved in E. coli invasion of HBMEC provides a novel approach for prevention of E. coli penetration into the brain, the essential step required for development of E. coli meningitis.


Subject(s)
Acetates/administration & dosage , Blood-Brain Barrier/microbiology , Endothelial Cells/microbiology , Escherichia coli/pathogenicity , Etoposide/administration & dosage , Meningitis, Escherichia coli/prevention & control , Quinolines/administration & dosage , Animals , Animals, Newborn , Brain/microbiology , Cells, Cultured , Cyclopropanes , Female , Humans , Leukotriene Antagonists , Rats , Rats, Sprague-Dawley , Receptors, Laminin/antagonists & inhibitors , Sulfides
6.
Infect Immun ; 78(7): 3090-6, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20421385

ABSTRACT

Escherichia coli K1 is the most common Gram-negative bacillary organism causing neonatal meningitis. E. coli K1 binding to and invasion of human brain microvascular endothelial cells (HBMECs) is a prerequisite for its traversal of the blood-brain barrier (BBB) and penetration into the brain. In the present study, we identified NlpI as a novel bacterial determinant contributing to E. coli K1 interaction with HBMECs. The deletion of nlpI did not affect the expression of the known bacterial determinants involved in E. coli K1-HBMEC interaction, such as type 1 fimbriae, flagella, and OmpA, and the contribution of NlpI to HBMECs binding and invasion was independent of those bacterial determinants. Previous reports have shown that the nlpI mutant of E. coli K-12 exhibits growth defect at 42 degrees C at low osmolarity, and its thermosensitive phenotype can be suppressed by a mutation on the spr gene. The nlpI mutant of strain RS218 exhibited similar thermosensitive phenotype, but additional spr mutation did not restore the ability of the nlpI mutant to interact with HBMECs. These findings suggest the decreased ability of the nlpI mutant to interact with HBMECs is not associated with the thermosensitive phenotype. NlpI was determined as an outer membrane-anchored protein in E. coli, and the nlpI mutant was defective in cytosolic phospholipase A(2)alpha (cPLA(2)alpha) phosphorylation compared to the parent strain. These findings illustrate the first demonstration of NlpI's contribution to E. coli K1 binding to and invasion of HBMECs, and its contribution is likely to involve cPLA(2)alpha.


Subject(s)
Bacterial Adhesion/physiology , Brain/microbiology , Endothelial Cells/microbiology , Escherichia coli Proteins/physiology , Escherichia coli/physiology , Lipoproteins/physiology , Meningitis, Escherichia coli/microbiology , Bacterial Outer Membrane Proteins/physiology , Blotting, Western , Brain/blood supply , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/physiology , Flagella/genetics , Flagella/physiology , Group IV Phospholipases A2/physiology , Humans , Infant, Newborn , Lipoproteins/genetics , Microcirculation
7.
Cell Tissue Res ; 332(3): 427-33, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18379824

ABSTRACT

Escherichia coli K1 invasion of microvascular endothelial cells of human brain (HBMEC) is required for E. coli penetration into the central nervous system, but the microbial-host interactions that are involved in this invasion of HBMEC remain incompletely understood. We have previously shown that FimH, one of the E. coli determinants contributing to the binding to and invasion of HBMEC, induces Ca(2+) changes in HBMEC. In the present study, we have investigated in detail the role of cellular calcium signaling in the E. coli K1 invasion of HBMEC, the main constituents of the blood-brain barrier. Addition of the meningitis-causing E. coli K1 strain RS218 (O18:K1) to HBMEC results in transient increases of intracellular free Ca(2+). Inhibition of phospholipase C with U-73122 and the chelating of intracellular Ca(2+) by BAPTA/AM reduces bacterial invasion of HBMEC by approximately 50%. Blocking of transmembrane Ca(2+) fluxes by extracellular lanthanum ions also inhibits the E. coli invasion of HBMEC by approximately 50%. In addition, E. coli K1 invasion is significantly inhibited when HBMEC are pretreated by the calmodulin antagonists, trifluoperazine or calmidazolium, or by ML-7, a specific inhibitor of Ca(2+)/calmodulin-dependent myosin light-chain kinase. These findings indicate that host intracellular Ca(2+) signaling contributes in part to E. coli K1 invasion of HBMEC.


Subject(s)
Brain/blood supply , Brain/microbiology , Calcium/metabolism , Calmodulin/metabolism , Endothelium, Vascular/microbiology , Escherichia coli/pathogenicity , Calcium Signaling , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Microcirculation/cytology
8.
Neurobiol Dis ; 27(3): 362-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17632007

ABSTRACT

Machado-Joseph disease also called spinocerebellar ataxia type 3 (MJD/SCA3) is a hereditary and neurodegenerative movement disorder caused by ataxin-3 with a polyglutamine expansion (mutant ataxin-3). Neuronal loss in MJD/SCA3 is associated with a mutant ataxin-3 toxic fragment. Defining mutant ataxin-3 proteolytic site(s) could facilitate the identification of the corresponding enzyme(s). Previously, we reported a mutant ataxin-3 mjd1a fragment in the brain of transgenic mice (Q71) that contained epitopes C-terminal to amino acid 220. In this study, we generated and characterized neuroblastoma cells and transgenic mice expressing mutant ataxin-3 mjd1a lacking amino acids 190-220 (deltaQ71). Less deltaQ71 than Q71 fragments were detected in the cell but not mouse model. The transgenic mice developed an MJD/SCA3-like phenotype and their brain homogenates had a fragment containing epitopes C-terminal to amino acid 220. Our results support the toxic fragment hypothesis and narrow the mutant ataxin-3 cleavage site to the N-terminus of amino acid 190.


Subject(s)
Brain/metabolism , Machado-Joseph Disease/genetics , Machado-Joseph Disease/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Animals , Ataxin-3 , Blotting, Western , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Mutation , Polymerase Chain Reaction , Transfection
9.
N J Med ; 101(10): 52; author reply 52-3, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15554441
10.
J Neuroimmunol ; 153(1-2): 150-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15265673

ABSTRACT

Chemokines and their receptors may be implicated in leukocyte ingress into brain during inflammation observed during the course of multiple sclerosis (MS). To address receptor modulation on CD4+ memory T lymphocytes during diapedesis, we used an in vitro model of the blood-brain barrier (BBB). We found that only memory (CD45RO+) cells transmigrated and type 3 CXC chemokine receptor (CXCR3) was enriched on transmigrated cells. CXCR3 depletion of the input population did not affect transmigration capability. CXCR3 reemerged on CXCR3 depleted cells independently of endothelial cell exposure, but was susceptible to incubation at 4 degrees C, indicating receptor recycling. We propose that CXCR3 serves as a surface marker for cells that have the capacity to cross the BBB, but does not play an essential role in extravasation.


Subject(s)
Brain/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Movement/physiology , Endothelium, Vascular/metabolism , Receptors, Chemokine/metabolism , Blotting, Western/methods , Brain/metabolism , Cell Line, Transformed , Chemokines/metabolism , Electric Impedance , Electrophoretic Mobility Shift Assay/methods , Flow Cytometry/methods , Humans , Immunohistochemistry/methods , Immunologic Memory , Membrane Proteins/metabolism , Occludin , Phosphoproteins/metabolism , Receptors, CXCR3 , Time Factors , Zonula Occludens-1 Protein
11.
Endothelium ; 11(5-6): 275-84, 2004.
Article in English | MEDLINE | ID: mdl-15770770

ABSTRACT

Central nervous system (CNS) dysfunction is commonly observed in children with human immunodeficiency virus type 1 (HIV-1) infection, but the mechanism(s) whereby HIV-1 causes encephalopathy remains incompletely understood. Human brain microvascular endothelial cells (HBMECs), which constitute the blood-brain barrier, are likely to contribute to HIV-1 encephalopathy, but it is unclear whether HIV-1 receptors (CD4, chemokine receptors) are present on HBMECs. In the present study, the presence of CD4 in six different children was demonstrated. Moreover, the presence of CD4 in situ on brain sections was shown. Distribution of CD4 expression was heterogeneous among microvessels; staining for CD4 was strong in some vessels and absent in other adjacent vessels. CD4 and chemokine coreceptors were found to be functional as intercellular adhesion molecule (ICAM)-1 expression increased upon incubation of HBMECs with activating anti-CD4 and anti-chemokine receptor antibodies. The presence of CD4 and chemokine receptors in human brain endothelium of children may have implications for the pathogenesis of HIV-1 encephalopathy and explain the higher incidence of CNS involvement in HIV-1-infected children as compared to adults.


Subject(s)
Brain/metabolism , CD4 Antigens/metabolism , Endothelial Cells/metabolism , HIV Infections/metabolism , HIV-1/metabolism , Receptors, Chemokine/metabolism , Animals , Endothelial Cells/virology , Endothelium, Vascular/metabolism , Endothelium, Vascular/virology , HIV Envelope Protein gp120/metabolism , HIV-1/pathogenicity , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Reverse Transcriptase Polymerase Chain Reaction
12.
Lab Invest ; 83(12): 1787-98, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14691297

ABSTRACT

Central nervous system dysfunction is commonly observed in children with HIV-1 infection, but the mechanisms whereby HIV-1 causes encephalopathy are not completely understood. We have previously shown that human brain microvascular endothelial cells (HBMEC) from children are responsive to gp120 derived from X4 HIV-1 by increasing expression of intercellular adhesion molecule (ICAM)-1 and vascular cell adhesion molecule-1. However, the mechanisms involved in gp120-mediated up-regulation of cell adhesion molecule expression is unclear. In the present study, we found that gp120 derived from both X4 and R5 HIV-1 induced increased expression of ICAM-1 on HBMEC, but the degree of this up-regulation differed among the various HBMEC isolates. The up-regulation of ICAM-1 was inhibited by anti-CD4 antibodies as well as by specific antibodies directed against chemokine receptors and small-molecule coreceptor inhibitors. Anti-CD4 antibodies inhibited the increase in ICAM-1 expression mediated by gp120 derived from X4 and R5 HIV-1, whereas antibodies against chemokine receptors displayed a differential inhibition depending on the source of gp120. Both X4 and R5 gp120-induced ICAM-1 expression was sensitive to pertussis toxin and involved the nuclear factor-kB pathway. These findings indicate a direct involvement of CD4 and a differential involvement of chemokine receptors in the activation of pediatric HBMEC by X4 and R5 gp120. The activation of brain endothelium of children by HIV-1 protein gp120 by way of CD4 and chemokine receptors may have implications for the pathogenesis of HIV-1 encephalopathy in the pediatric population.


Subject(s)
CD4 Antigens/metabolism , Endothelium, Vascular/metabolism , HIV Envelope Protein gp120/pharmacology , HIV-1 , Intercellular Adhesion Molecule-1/biosynthesis , Nitriles , Sulfones , Antibodies, Blocking/pharmacology , CD4 Antigens/immunology , Cells, Cultured , Child , Endothelium, Vascular/drug effects , Humans , Intercellular Adhesion Molecule-1/genetics , Microcirculation/cytology , NF-kappa B/antagonists & inhibitors , Organic Chemicals/pharmacology , Pertussis Toxin/pharmacology , RNA, Messenger/metabolism , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...