Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Infect Dis ; 184(6): 732-7, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11517434

RESUMO

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.


Assuntos
Benzilisoquinolinas , Circulação Cerebrovascular/fisiologia , Endotélio Vascular/enzimologia , Endotélio Vascular/microbiologia , Escherichia coli/patogenicidade , Listeria monocytogenes/patogenicidade , Fosfolipases A/metabolismo , Alcaloides/farmacologia , Ácidos Araquidônicos/farmacologia , Células Cultivadas , Córtex Cerebral/irrigação sanguínea , Criança , Pré-Escolar , Citosol/enzimologia , Medicamentos de Ervas Chinesas/farmacologia , Inibidores Enzimáticos/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/fisiologia , Humanos , Indometacina/farmacologia , Inibidores de Lipoxigenase/farmacologia , Listeria monocytogenes/efeitos dos fármacos , Listeria monocytogenes/fisiologia , Masoprocol/farmacologia , Microcirculação/enzimologia , Microcirculação/microbiologia , Fosfolipases A/antagonistas & inibidores , Fosfolipases A2 , Transdução de Sinais
2.
Infect Immun ; 68(11): 6423-30, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11035755

RESUMO

Escherichia coli K1 traversal across the blood-brain barrier is an essential step in the pathogenesis of neonatal meningitis. We have previously shown that invasive E. coli promotes the actin rearrangement of brain microvascular endothelial cells (BMEC), which constitute a lining of the blood-brain barrier, for invasion. However, signal transduction mechanisms involved in E. coli invasion are not defined. In this report we show that tyrosine kinases play a major role in E. coli invasion of human BMEC (HBMEC). E. coli induced tyrosine phosphorylation of HBMEC cytoskeletal proteins, focal adhesion kinase (FAK), and paxillin, with a concomitant increase in the association of paxillin with FAK. Overexpression of a dominant interfering form of the FAK C-terminal domain, FRNK (FAK-related nonkinase), significantly inhibited E. coli invasion of HBMEC. Furthermore, we found that FAK kinase activity and the autophosphorylation site (Tyr397) are important in E. coli invasion of HBMEC, whereas the Grb2 binding site (Tyr925) is not required. Immunocytochemical studies demonstrated that FAK is recruited to focal plaques at the site of bacterial entry. Consistent with the invasion results, overexpression of FRNK, a kinase-negative mutant (Arg454 FAK), and a Src binding mutant (Phe397 FAK) inhibited the accumulation of FAK at the bacterial entry site. The overexpression of FAK mutants in HBMEC also blocked the E. coli-induced tyrosine phosphorylation of FAK and its association with paxillin. These observations provide evidence that FAK tyrosine phosphorylation and its recruitment to the cytoskeleton play a key role in E. coli invasion of HBMEC.


Assuntos
Encéfalo/microbiologia , Endotélio Vascular/microbiologia , Escherichia coli/patogenicidade , Proteínas Tirosina Quinases/fisiologia , Encéfalo/irrigação sanguínea , Células Cultivadas , Ativação Enzimática , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Genisteína/farmacologia , Humanos , Fosforilação , Tirosina/metabolismo
3.
J Biol Chem ; 275(47): 36769-74, 2000 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-10973983

RESUMO

Invasion of brain microvascular endothelial cells (BMEC) is a prerequisite for successful crossing of the blood-brain barrier by Escherichia coli K1. We have previously demonstrated the requirement of cytoskeletal rearrangements and activation of focal adhesion kinase (FAK) in E. coli K1 invasion of human BMEC (HBMEC). The current study investigated the role of phosphatidylinositol 3-kinase (PI3K) activation and PI3K interaction with FAK in E. coli invasion of HBMEC. PI3K inhibitor LY294002 blocked E. coli K1 invasion of HBMEC in a dose-dependent manner, whereas an inactive analogue LY303511 had no such effect. In HBMEC, E. coli K1 increased phosphorylation of Akt, a downstream effector of PI3K, which was completely blocked by LY294002. In contrast, non-invasive E. coli failed to activate PI3K. Overexpression of PI3K mutants Deltap85 and catalytically inactive p110 in HBMEC significantly inhibited both PI3K/Akt activation and E. coli K1 invasion of HBMEC. Stimulation of HBMEC with E. coli K1 increased PI3K association with FAK. Furthermore, PI3K/Akt activation was blocked in HBMEC-overexpressing FAK dominant-negative mutants (FRNK and Phe397FAK). These results demonstrated the involvement of PI3K signaling in E. coli K1 invasion of HBMEC and identified a novel role for PI3K interaction with FAK in the pathogenesis of E. coli meningitis.


Assuntos
Encéfalo/microbiologia , Circulação Cerebrovascular , Endotélio Vascular/enzimologia , Endotélio Vascular/microbiologia , Escherichia coli/patogenicidade , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Barreira Hematoencefálica , Células Cultivadas , Cromonas/farmacologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Escherichia coli/enzimologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Microcirculação , Morfolinas/farmacologia , Mutagênese Sítio-Dirigida , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Transdução de Sinais , Relação Estrutura-Atividade
4.
Infect Immun ; 68(9): 5056-61, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10948125

RESUMO

Escherichia coli K1 is the leading cause of gram-negative bacterial meningitis in neonates. It is principally due to our limited understanding of the pathogenesis of this disease that the morbidity and mortality rates remain unacceptably high. To identify genes required for E. coli K1 penetration of the blood-brain barrier (BBB), we used the negative selection strategy of signature-tagged transposon mutagenesis (STM) to screen mutants for loss or decreased invasion of human brain microvascular endothelial cells (HBMEC) which comprise the BBB. A total of 3,360 insertion mutants of E. coli K1 were screened, and potential HBMEC invasion mutants were subjected to a secondary invasion screen. Those mutants that failed to pass the serial invasion screens were then tested individually. Seven prototrophic mutants were found to exhibit significantly decreased invasive ability in HBMEC. We identified traJ and five previously uncharacterized loci whose gene products are necessary for HBMEC invasion by E. coli K1. In addition, cnf1, a gene previously shown to play a role in bacterial invasion, was identified. More importantly, a traJ mutant was attenuated in penetration of the BBB in the neonatal rat model of experimental hematogenous meningitis. This is the first in vivo demonstration that traJ is involved in the pathogenesis of E. coli K1 meningitis.


Assuntos
Encéfalo/microbiologia , Endotélio Vascular/microbiologia , Escherichia coli/genética , Genes Bacterianos , Animais , Barreira Hematoencefálica , Encéfalo/irrigação sanguínea , Elementos de DNA Transponíveis , Endotélio Vascular/citologia , Escherichia coli/patogenicidade , Humanos , Meningites Bacterianas/etiologia , Mutagênese , Ratos
5.
Mol Microbiol ; 36(1): 174-82, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10760174

RESUMO

Most cases of Escherichia coli K1 meningitis arise as a result of haematogenous spread, however there is a limited understanding of the mechanisms by which circulating E. coli K1 cross the blood-brain barrier. We have previously shown that environmental growth conditions both positively and negatively influence the capabilities of E. coli K1 to invade brain microvascular endothelial cells (BMEC), for example growth in media supplemented with 50% newborn bovine serum (NBS) increased BMEC invasion, whereas growth in media supplemented with 0.2 M NaCl repressed invasion in vitro and in vivo. In this study, differential fluorescence induction (DFI) was used to identify E. coli K1 genes involved in this differentially expressed invasion phenotype. E. coli K1 promoter libraries were constructed and screened for gfp expression in a manner analogous to the above growth conditions. Twenty-four clones were isolated that showed fluorescence induction when grown under the invasion-enhancing condition (i.e. NBS). Four of these clones also demonstrated repression or no induction of fluorescence when grown under the invasion-repressing condition (i.e. 0.2 M NaCl). One such clone, containing a ygdP promoter and an open reading frame (ORF), showed significant homology to Bartonella bacilliformis IalA (invasion associated locus). Among the other NBS-inducing loci, finPtraJ was identified as well as several clones with no homology to other known genes. When ygdP, finPtraJ and several of the unique loci were disrupted in E. coli K1, there was a significant decrease in human BMEC (HBMEC) invasion. RNA transcript analysis determined that these newly identified invasion loci were differentially regulated at the transcriptional level. This is the first demonstration of using DFI to identify E. coli K1 genes contributing to HBMEC invasion.


Assuntos
Encéfalo/irrigação sanguínea , Endotélio Vascular/microbiologia , Escherichia coli/genética , Escherichia coli/patogenicidade , Meningite devida a Escherichia coli/microbiologia , Encéfalo/citologia , Células Cultivadas , Clonagem Molecular , Meios de Cultura , Endotélio Vascular/citologia , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Teste de Complementação Genética , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Mutagênese , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão , Análise de Sequência de DNA
6.
Infect Immun ; 67(11): 5775-83, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10531228

RESUMO

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.


Assuntos
Actinas/metabolismo , Proteínas da Membrana Bacteriana Externa/fisiologia , Encéfalo/microbiologia , Endotélio Vascular/microbiologia , Escherichia coli/fisiologia , Animais , Encéfalo/irrigação sanguínea , Bovinos , Clatrina/fisiologia , Citocalasina D/farmacologia , Endocitose , Endotélio Vascular/ultraestrutura , Microscopia Eletrônica , Microtúbulos/fisiologia
7.
Infect Immun ; 67(9): 4751-6, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10456927

RESUMO

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.


Assuntos
Proteínas de Bactérias/genética , Encéfalo/microbiologia , Endotélio Vascular/microbiologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli , Escherichia coli/genética , Proteínas de Membrana , Meningites Bacterianas/microbiologia , Animais , Proteínas de Bactérias/fisiologia , Sequência de Bases , Barreira Hematoencefálica , Encéfalo/irrigação sanguínea , Bovinos , Deleção Cromossômica , Mapeamento Cromossômico , Cromossomos Bacterianos , Clonagem Molecular , DNA Bacteriano , Endotélio Vascular/citologia , Escherichia coli/isolamento & purificação , Escherichia coli/patogenicidade , Genes Bacterianos , Teste de Complementação Genética , Humanos , Dados de Sequência Molecular , Mutagênese Insercional , Ratos
8.
Infect Immun ; 67(8): 4153-60, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10417187

RESUMO

Opacity-associated protein A (OapA), which is responsible for the transparent-colony phenotype of Haemophilus influenzae, has been implicated in the colonization of the nasopharynx in an infant rat model of carriage. In this report, we show that OapA mediates attachment to Chang epithelial cells examined by using genetically defined type b and nontypeable H. influenzae strains with or without OapA. We also showed that OapA was conserved among H. influenzae strains by comparing deduced amino acid sequences. Both recombinant OapA and polyclonal anti-OapA antiserum blocked the binding of H. influenzae to Chang epithelial cells, suggesting that the interaction of H. influenzae is specific to OapA. Moreover, the binding of recombinant OapA to epithelial cells further provided evidence that OapA can promote attachment of H. influenzae. Expression of oapA gene in a nonadherent Escherichia coli strain significantly increased the binding to Chang epithelial cells, and disruption of the oapA gene with kanamycin resistance cassette insertion resulted in a significant loss of binding. These findings demonstrate that OapA plays a role in H. influenzae binding to human conjunctival epithelial cells.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/fisiologia , Túnica Conjuntiva/microbiologia , Haemophilus influenzae/fisiologia , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Células Epiteliais/microbiologia , Humanos , Dados de Sequência Molecular , Nasofaringe/microbiologia , Ratos
9.
Infect Immun ; 67(3): 1131-8, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10024553

RESUMO

The molecular basis of Escherichia coli traversal of the blood-brain barrier in the development of E. coli meningitis is not well understood. We have previously shown that a novel Ibe10 protein found in cerebrospinal fluid isolates of E. coli is necessary for invasion of the brain microvascular endothelial cells (BMEC) that constitute the blood-brain barrier both in vitro and in a newborn rat model of hematogenous meningitis. Here we identified a novel Ibe10 binding molecule/receptor (Ibe10R) on both bovine BMEC (HBMEC) and human BMEC (HBMEC) that is responsible for invasion by E. coli. Ibe10R, an approximately 55-kDa protein, was purified from BBMEC by Ibe10-Ni-Sepharose affinity chromatography. Bovine Ibe10R, as well as polyclonal antibodies to Ibe10R, blocked E. coli invasion of BBMEC very effectively. The N-terminal amino acid sequence of Ibe10R showed 75% homology to serum albumin. However, the amino acid sequence of an Ibe10R fragment generated by limited enzymatic digestion did not reveal homology to any other proteins, suggesting that Ibe10R represents a novel albumin-like protein. Immunocytochemical analysis of BBMEC using anti-Ibe10R antibody suggested that only a subset of cultured BBMEC express Ibe10R on their surface. Enrichment of Ibe10R-positive BBMEC by fluorescence-activated cell sorting with anti-Ibe10R antibody resulted in enhanced invasion by E. coli. The anti-Ibe10R antibody raised against bovine Ibe10R also blocked E. coli invasion of HBMEC very effectively. Interestingly, anti-Ibe10R antibody affinity chromatography of HBMEC membrane proteins revealed a smaller protein with an approximate molecular mass of 45 kDa. These results suggest that the Ibe10 of E. coli interacts with a novel BMEC surface protein, Ibe10R, for invasion of both BBMEC and HBMEC.


Assuntos
Proteínas de Bactérias/fisiologia , Encéfalo/microbiologia , Proteínas de Transporte/fisiologia , Endotélio Vascular/microbiologia , Proteínas de Escherichia coli , Escherichia coli/patogenicidade , Proteínas de Membrana/fisiologia , Animais , Proteínas de Transporte/análise , Bovinos , Humanos , Recém-Nascido , Ratos
10.
Infect Immun ; 65(7): 2852-60, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9199459

RESUMO

We have previously shown that S-fimbriated Escherichia coli binds brain microvascular endothelial cells (BMEC) via a lectin-like activity of SfaS adhesin specific for NeuAc alpha2,3-galactose; however, BMEC molecules bearing these epitopes have not been identified. In the present study, we showed that the expression of S fimbriae conferred a three-fold increase in adhesion of E. coli to cow, human, and rat BMEC but did not enhance E. coli adhesion to systemic vascular endothelial cells such as human umbilical vein endothelial cells and human aortic arterial endothelial cells. Two BMEC-binding molecules for S fimbriae were identified as 65 (major)- and 130 (minor)-kDa sialoglycoproteins by S fimbria immunoblotting and were purified from bovine BMEC by wheat germ agglutinin and Maackia amurensis lectin (specific to NeuAc alpha2,3-galactose) affinity chromatography. The 65-kDa BMEC glycoprotein showed effective inhibition of S fimbria-mediated binding of E. coli to BMEC. Polyclonal antibodies raised against the mixture of 65- and 130-kDa proteins reacted to 65-kDa protein present only on BMEC, not on systemic vascular endothelial cells. Immunoprecipitation of biotinylated BMEC membrane proteins and immunocytochemistry studies of BMEC with anti-S fimbria-binding protein antibodies revealed that the 65-kDa protein is a surface protein. The N-terminal amino acid sequence of 65- and 130-kDa proteins showed no significant sequence homology with any other known proteins. These findings suggest that 65- and 130-kDa proteins represent novel sialoglycoproteins involved in the binding of S-fimbriated E. coli to BMEC.


Assuntos
Circulação Cerebrovascular , Endotélio Vascular/fisiologia , Fímbrias Bacterianas/imunologia , Sialoglicoproteínas/imunologia , Sialoglicoproteínas/isolamento & purificação , Animais , Bovinos , Escherichia coli , Fímbrias Bacterianas/fisiologia , Humanos , Ratos
11.
Exp Neurol ; 145(1): 253-7, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9184127

RESUMO

In an attempt to examine whether routes of bacterial entry into the central nervous system have any bearing on subsequent changes in blood-brain barrier permeability, we examined cerebrospinal fluid (CSF) penetration of circulating 125I-albumin in two different models of experimental meningitis due to K1 Escherichia coli, type III group B streptococcus, or Haemophilus influenzae type b in infant rats: hematogenous meningitis subsequent to subcutaneous inoculation of bacteria vs meningitis induced by direct inoculation of bacteria into the CSF via the cisterna magna. In the model of hematogenous meningitis, the mean CSF penetration was significantly greater in animals with H. influenzae type b meningitis than in those with meningitis due to K1 E. coli or type III group B streptococcus. In contrast, the mean CSF penetration was significantly enhanced in all animals with meningitis induced by intracisternal inoculation regardless of infecting pathogens. Tumor necrosis factor activity in CSF appeared to correlate with the functional penetration of circulating albumin across the blood-brain barrier in both models of experimental meningitis. These findings suggest that the alterations of blood-brain barrier permeability during development of experimental meningitis may vary for different models of inducing meningitis and that the mechanisms responsible for these different permeability changes may be multifactorial.


Assuntos
Barreira Hematoencefálica/fisiologia , Meningite por Haemophilus/sangue , Soroalbumina Bovina/farmacocinética , Streptococcus agalactiae , Animais , Bacteriemia/metabolismo , Permeabilidade da Membrana Celular , Modelos Animais de Doenças , Infecções por Escherichia coli/sangue , Feminino , Injeções Espinhais , Radioisótopos do Iodo , Gravidez , Ratos , Ratos Sprague-Dawley , Organismos Livres de Patógenos Específicos , Infecções Estreptocócicas/sangue , Fator de Necrose Tumoral alfa/líquido cefalorraquidiano
12.
Infect Immun ; 64(1): 146-53, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8557332

RESUMO

Escherichia coli is the most common gram-negative bacteria causing meningitis during the neonatal period, but is unclear what microbial factors mediate traversal of E. coli across the blood-brain barrier. Outer membrane protein A (OmpA), a highly conserved 35-kDa protein, was examined for its role in E. coli K1 invasion of brain microvascular endothelial cells (BMEC). The invasive capability of the OmpA+ strains was 25- to 50-fold greater than that of OmpA- strains, and the invasive capability of OmpA- strains was restored to the level of the OmpA+ strain by complementation with the OmpA+ E. coli into BMEC. Two short synthetic peptides (a hexamer, Asn-27-Glu-32, and a pentamer, Gly-65-Asn-69) generated from the N-terminal amino acid sequence of OmpA exhibited significant inhibition of OmpA+ E. coli invasion, suggesting that these two sequences represent the OmpA domains involved in E. coli invasion of BMEC. These findings suggest that OmpA is the first microbial structure identified to enhance E. coli invasion of BMEC, an important event in the pathogenesis of E. coli meningitis.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Encéfalo/microbiologia , Capilares/microbiologia , Endotélio Vascular/microbiologia , Escherichia coli/patogenicidade , Sequência de Aminoácidos , Animais , Anticorpos Antibacterianos/farmacologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Capilares/citologia , Bovinos , Escherichia coli/genética , Teste de Complementação Genética , Técnicas In Vitro , Dados de Sequência Molecular , Mutação , Oligopeptídeos/farmacologia , Virulência/efeitos dos fármacos , Virulência/genética
13.
Infect Immun ; 64(1): 154-60, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8557333

RESUMO

Inadequate knowledge of pathogenesis and pathophysiology has contributed to the high mortality and morbidity associated with neonatal Escherichia coli meningitis. We have shown previously that outer membrane protein A (OmpA) contributes to E. coli K1 membrane invasion of brain microvascular endothelial cells. In this study we report that this OmpA+ K1 E. coli invasion of brain microvascular endothelial cells was inhibited by wheat germ agglutinin and chitooligomers prepared from the polymer of 1,4-linked GlcNAc, chitin. The specificity of the interaction between OmpA and GlcNAc beta 1-4GlcNAc epitopes was verified by the demonstration that chitotriose-bound OmpA and wheat germ agglutinin-bound brain microvascular endothelial cell membrane proteins inhibit E. coli K1 invasion. Of interest, OmpA+ E. coli invasion into systemic endothelial cells did not occur, but invasion similar to that of brain microvascular endothelial cells was observed when systemic cells were treated with alpha-fucosidase, suggesting that the GlcNAc beta 1-4GlcNAc moieties might be substituted with L-fucose on these cells. More importantly, the chitooligomers prevented entry of E. coli K1 into the cerebrospinal fluid of newborn rats with experimental hematogenous E. coli meningitis, suggesting that the GlcNAc beta 1-4GlcNAc epitope of brain microvascular endothelial cells indeed mediates the traversal of E. coli K1 across the blood-brain barrier. A novel strategy with the use of soluble receptor analog(s) may be feasible in the prevention of devastating neonatal E. coli meningitis.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Barreira Hematoencefálica , Endotélio Vascular/microbiologia , Escherichia coli/patogenicidade , Maltose/análogos & derivados , Animais , Sequência de Carboidratos , Carboidratos/farmacologia , Bovinos , Sistema Nervoso Central/microbiologia , Líquido Cefalorraquidiano/microbiologia , Endotélio Vascular/química , Epitopos , Infecções por Escherichia coli/etiologia , Infecções por Escherichia coli/microbiologia , Glicoproteínas/farmacologia , Maltose/metabolismo , Meningites Bacterianas/etiologia , Meningites Bacterianas/microbiologia , Dados de Sequência Molecular , Ratos , Ratos Sprague-Dawley , Virulência/efeitos dos fármacos , Aglutininas do Germe de Trigo/farmacologia , alfa-L-Fucosidase/farmacologia
14.
Am J Pathol ; 145(5): 1228-36, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7977653

RESUMO

To assess the role of S fimbriae in the pathogenesis of Escherichia coli meningitis, transformants of E. coli strains with or without S fimbriae plasmid were compared for their binding to microvessel endothelial cells isolated from bovine brain cortices (BMEC). The BMEC's displayed a cobblestone appearance, were positive for factor VIII, carbonic anhydrase IV, took up fluorescent-labeled acetylated low density lipoprotein, and exhibited gamma glutamyl transpeptidase activity. Binding of S fimbriated E. coli to BMEC was approximately threefold greater than nonfimbriated E. coli Similarly S fimbriated E. coli bound to human brain endothelial cells approximately threefold greater than nonfimbriated E. coli. Binding was reduced approximately 60% by isolated S fimbriae and about 80% by anti-S adhesin antibody. Mutating the S adhesin gene resulted in a complete loss of the binding, whereas mutagenesis of the major S fimbriae subunit gene sfaA did not significantly affect binding. Pretreatment of BMEC with neuraminidase or prior incubation of S fimbriated E. coli with NeuAc alpha 2,3-sialyl lactose completely abolished binding. These findings indicate that S fimbriated E. coli bind to NeuAc alpha 2,3-galactose containing glycoproteins on brain endothelial cells via a lectin-like activity of SfaS adhesin. This might be an important early step in the penetration of bacteria across the blood-brain barrier in the development of E. coli meningitis.


Assuntos
Aderência Bacteriana/fisiologia , Encéfalo/metabolismo , Endotélio Vascular/metabolismo , Escherichia coli/metabolismo , Fímbrias Bacterianas/metabolismo , Animais , Bovinos , Células Cultivadas , Endotélio Vascular/citologia , Escherichia coli/genética , Humanos , Lactose/análogos & derivados , Lactose/metabolismo , Mutação , Ácidos Siálicos/metabolismo
15.
J Biol Chem ; 268(14): 10356-63, 1993 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8098039

RESUMO

In an attempt to further assess the role of S-fimbriae in the pathogenesis of Escherichia coli meningitis, the adherence of E. coli strains with or without S-fimbriae were examined for this study to purified glycolipids using thin layer chromatography overlay assays. Only S-fimbriated E. coli strains bound to sulfatide, seminolipid, galactosyl ceramide, and lactosyl ceramide but not to gangliosides including sialyl neolacto-series and other neutral glycolipids. The binding of S-fimbriated E. coli to sulfatide was temperature dependent (i.e. maximal at 37 degrees C) and inhibited by S-fimbriae, anti-S-fimbriae, and anti-S-adhesin antibodies as well as by sulfatide, galactosyl ceramide, and lactosyl ceramide. E. coli transformants which lack the sfaA gene from the Sfa gene cluster showed no binding to the glycolipids, while other transformants lacking the adhesin gene sfaS or sfaG or H and mutants obtained by site-directed mutagenesis in the sfaS gene exhibited a similar binding to the glycolipids compared to the parent S-fimbriated strain. A large amount of sulfated glycolipids was demonstrated on brain endothelial cells and the binding of S-fimbriated E. coli to brain endothelial cells was inhibited by these glycolipids. These findings suggest that the binding of S-fimbriated E. coli to brain endothelial cells occurs in part via glycolipids containing terminal Gal(3SO4)beta-1 residues and in part by S-fimbriae protein SfaA. S-adhesin was not involved in the binding of S-fimbriae to these glycolipids.


Assuntos
Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/metabolismo , Encéfalo/metabolismo , Escherichia coli/fisiologia , Fímbrias Bacterianas/fisiologia , Gangliosídeos/metabolismo , Genes Bacterianos , Glicolipídeos/metabolismo , Adesinas de Escherichia coli , Animais , Proteínas da Membrana Bacteriana Externa/genética , Configuração de Carboidratos , Sequência de Carboidratos , Bovinos , Circulação Cerebrovascular , Endotélio Vascular/metabolismo , Escherichia coli/genética , Gangliosídeos/isolamento & purificação , Genótipo , Cinética , Dados de Sequência Molecular , Fenótipo , Termodinâmica
16.
Infect Immun ; 61(3): 975-80, 1993 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7679376

RESUMO

Serum obtained after immunization with an O18 polysaccharide-toxin A conjugate vaccine was evaluated for the estimation of protective levels of anti-O-specific lipopolysaccharide (LPS) immunoglobulin G (IgG) antibody against bacteremia and death caused by a homologous serotype of Escherichia coli K1 strains. Passive transfer of rabbit serum conferred significant protection from a lethal E. coli infection in a neonatal rat model. The overall incidence of bacteremia and mortality was 4% in rat pups receiving undiluted postvaccination serum, while that in control animals was 100% (P < 0.001). The overall incidences of bacteremia were 5 and 72% for animals with serum anti-O18 LPS IgG concentrations of > 1.0 and < 1.0 microgram/ml, respectively, while the overall incidences of mortality for animals with serum anti-O18 LPS IgG levels of > 1.0 and < 1.0 microgram/ml were 0 and 72%, respectively (P < 0.001). Protection against E. coli infection was also demonstrated with human anti-O18 polysaccharide IgG. None of the animals with human anti-O18 LPS IgG levels of > 1 microgram/ml had bacteremia after bacterial challenge, whereas all animals with bacteremia at 18 h had levels of < 1 microgram/ml. These findings suggest that serum anti-O18 LPS IgG concentrations of > 1.0 microgram/ml may provide protection against bacteremia and death caused by a homologous E. coli K1 infection.


Assuntos
Infecções por Escherichia coli/prevenção & controle , Imunoglobulina G/imunologia , Lipopolissacarídeos/imunologia , Polissacarídeos Bacterianos/imunologia , Animais , Bacteriemia/imunologia , Escherichia coli/imunologia , Infecções por Escherichia coli/imunologia , Feminino , Humanos , Antígenos O , Gravidez , Coelhos , Ratos , Ratos Sprague-Dawley
17.
Lymphokine Cytokine Res ; 11(6): 293-8, 1992 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1477182

RESUMO

In an attempt to understand the role of TNF in the central nervous system (CNS) pathophysiologic events associated with bacterial meningitis, we examined the effect of intravenous vs. intracisternal administration of TNF alpha on penetration of circulating 125I-labeled albumin into cerebrospinal fluid (CSF) and CSF white blood cell (WBC) counts in rats. Intracisternal administration of tumor necrosis factor alpha (TNF-alpha) resulted in dose- and time-dependent alterations of the CSF penetration and CSF WBCs, while intravenous administration of TNF-alpha did not induce any changes. These changes by intracisternal TNF were abolished by heat treatment of TNF or coadministration of MAb to TNF-alpha. Mab to TNF-alpha also significantly reduced the CSF penetration of circulating albumin in experimental hematogenous Haemophilus influenzae type b meningitis in infant rats but this salutary effect required both intravenous and intracisternal administration. However, MAb to TNF-alpha failed to affect CSF pleocytosis in experimental hematogenous meningitis. These findings suggest that some of CNS pathophysiologic changes in bacterial meningitis may be a result of the local production of TNF but other host inflammatory responses may also participate in CNS inflammation in hematogenous bacterial meningitis.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/administração & dosagem , Animais , Anticorpos Monoclonais/administração & dosagem , Barreira Hematoencefálica/fisiologia , Líquido Cefalorraquidiano/citologia , Injeções Intraventriculares , Contagem de Leucócitos , Meningite por Haemophilus/líquido cefalorraquidiano , Meningite por Haemophilus/fisiopatologia , Permeabilidade/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Albumina Sérica/líquido cefalorraquidiano , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia
18.
J Clin Immunol ; 10(3): 154-9, 1990 May.
Artigo em Inglês | MEDLINE | ID: mdl-2195055

RESUMO

Preparations of IgG 3 isolated by absorption of IgG 1, IgG 2, and IgG 4 from a human iv immunoglobulin with protein A-Sepharose were evaluated for their opsonic activities against type III group B streptococcal (GBS) strains. The resulting preparations were free of IgG 1 and IgG 2 and contained only trace amounts of IgG 4 (less than 2% of total IgG). These IgG 3 preparations exhibited excellent opsonic activities against type III GBS strains, similar to those of the unfractionated iv immunoglobulin (based on total IgG concentrations in the opsonic assays). In contrast, preparations of IgG 1, 2, and 4 eluted from protein A-Sepharose with 2 M acetic acid and 7 M urea were significantly less effective in enhancing phagocytosis and killing of type III GBS than IgG 3 preparations or iv immunoglobulin. The reasons for excellent opsonic activity of IgG 3 preparations as well as for decreased opsonic activity of IgG 1, 2, and 4 preparations are not clear. Perhaps alteration of IgG by lower pH and high concentrations of urea may have impaired the functional activity of IgG 1, 2, and 4 preparations. The significant finding of this study is the first demonstration of the excellent opsonic activity of IgG 3, emphasizing the importance of having intact IgG 3 in commercial immunoglobulin preparations used in prophylaxis or treatment of GBS infections.


Assuntos
Imunoglobulina G/fisiologia , Proteínas Opsonizantes , Streptococcus agalactiae/imunologia , Humanos , Concentração de Íons de Hidrogênio , Imunoglobulina G/isolamento & purificação , Sefarose/análogos & derivados , Ureia
19.
Pediatr Res ; 24(5): 628-32, 1988 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-3060825

RESUMO

In an effort to further understand the host defense against group B streptococcus (GBS), we examined 71 human cord sera for their content of type III GBS IgG antibody by enzyme-linked immunosorbent assay and correlated the results with opsonic and protective activity against type III GBS. Most cord sera (67%) containing greater than 0.1 microgram/ml of type III GBS IgG antibody promoted phagocytosis and killing in vitro and protection against type III GBS in neonatal rats. However, 26% of cord sera containing less than 0.1 microgram/ml of type III IgG antibody exhibited similar activity in vitro and in vivo against type III GBS. This opsonic and protective activity was retained in IgG fraction of whole serum, and was not directly associated with complement activity or with fibronectin. Further studies are needed to understand the mechanisms responsible for the opsonic and protective activity of some cord sera against type III GBS that may be independent of antibody to the type-specific polysaccharide antigen.


Assuntos
Sangue Fetal/imunologia , Proteínas Opsonizantes , Streptococcus agalactiae/imunologia , Animais , Anticorpos Antibacterianos/análise , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Recém-Nascido , Gravidez , Ratos , Ratos Endogâmicos
20.
J Infect Dis ; 153(6): 1092-7, 1986 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-3517187

RESUMO

Two preparations of human immunoglobulin modified for intravenous use (iv immunoglobulin) by different methods (reduction-and-alkylation, or pH 4 treatment) were evaluated for in vitro and in vivo activity against a strain of type III group B Streptococcus (GBS). Both preparations contained similar amounts of total IgG and specific IgG antibody against the type-specific polysaccharide. In vitro, opsonophagocytic studies revealed that pH 4-treated iv immunoglobulin was significantly more effective than reduced-and-alkylated iv immunoglobulin in supporting neutrophil-mediated killing of the type III GBS strain. In vivo, both preparations resulted in similar levels of serum antibody in newborn rats, but pH 4-treated iv immunoglobulin was significantly more protective against the type III GBS strain. This was demonstrated by the lower magnitude of bacteremia, improved survival, and lower protective dose (PD50) in recipients of pH 4-treated iv immunoglobulin. Thus, reduced-and-alkylated iv immunoglobulin was less effective in vitro and in vivo against the strain of type III GBS than was pH 4-treated iv immunoglobulin. Further studies are needed to elucidate the mechanisms for this apparent discrepancy in functional activities of iv immunoglobulin.


Assuntos
Anticorpos Antibacterianos/imunologia , Infecções Estreptocócicas/prevenção & controle , Streptococcus agalactiae/imunologia , Alquilação , Animais , Animais Recém-Nascidos , Feminino , Humanos , Concentração de Íons de Hidrogênio , Imunização Passiva , Imunoglobulina G/classificação , Imunoglobulina G/imunologia , Técnicas In Vitro , Proteínas Opsonizantes , Oxirredução , Fagocitose , Gravidez , Ratos , Sepse/terapia , Infecções Estreptocócicas/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...