Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Epidemiol Infect ; 145(3): 600-606, 2017 02.
Article in English | MEDLINE | ID: mdl-27873565

ABSTRACT

Achromobacter spp. are opportunistic pathogens increasingly recovered from adult patients with cystic fibrosis (CF). We report the characterization of 122 Achromobacter spp. isolates recovered from 39 CF patients by multilocus sequence typing, virulence traits, and susceptibility to antimicrobials. Two species, A. xylosoxidans (77%) and A. ruhlandii (23%) were identified. All isolates showed a similar biofilm formation ability, and a positive swimming phenotype. By contrast, 4·3% and 44·4% of A. xylosoxidans and A. ruhlandii, respectively, exhibited a negative swarming phenotype, making the swimming and swarming abilities of A. xylosoxidans significantly higher than those of A. ruhlandii. A. xylosoxidans isolates from an outbreak clone also exhibited significantly higher motility. Both species were generally susceptible to ceftazidime, ciprofloxacin, imipenem and trimethoprim/sulphamethoxazole and there was no significant difference in susceptibility between isolates from chronic or sporadic infection. However, A. xylosoxidans isolates from chronic and sporadic cases were significantly more resistant to imipenem and ceftazidime than isolates of the outbreak clone.


Subject(s)
Achromobacter/isolation & purification , Cystic Fibrosis/complications , Gram-Negative Bacterial Infections/microbiology , Virulence Factors/analysis , Achromobacter/classification , Achromobacter/drug effects , Achromobacter/physiology , Anti-Bacterial Agents/pharmacology , Biofilms/growth & development , Humans , Locomotion , Microbial Sensitivity Tests , Multilocus Sequence Typing
2.
Eur Respir J ; 32(6): 1591-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18684846

ABSTRACT

The present study addressed the question whether ExoU, a Pseudomonas aeruginosa toxin with phospholipase A2 (PLA2) activity, may induce airway epithelial cells to overexpress tissue factor (TF) and exhibit a procoagulant phenotype. Cells from the human bronchial epithelial BEAS-2B line were infected with an ExoU-producing P. aeruginosa strain, pre-treated or not with the cytosolic PLA2 inhibitor methylarachidonyl fluorophosphate (MAFP), or with two ExoU-deficient mutants. Control noninfected and infected cells were assessed for the expression of: 1) TF mRNA by RT-PCR; 2) cell-associated TF by enzyme immunoassay and flow cytometry; 3) procoagulant activity by a colorimetric assay; and 4) microparticle-associated TF by flow cytometry. An enzyme immunoassay was also used to assess cell-associated TF in lung extracts from mice infected intratracheally with ExoU-producing and -deficient bacteria. Cells infected with the wild-type bacteria had higher levels of TF mRNA, cell-associated TF expression, procoagulant activity and released microparticle-associated TF than cells infected with the mutants. Bacterial treatment with MAFP significantly reduced the expression of TF by infected cells. Lung samples from mice infected with the wild-type bacteria exhibited higher levels of cell-associated TF and procoagulant activity. The present results demonstrate that ExoU may contribute to the pathogenesis of lung injury by inducing a tissue factor-dependent procoagulant activity in airway epithelial cells.


Subject(s)
Bacterial Proteins/physiology , Bronchi/microbiology , Coagulants/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/metabolism , Animals , Arachidonic Acids/metabolism , Bacterial Proteins/metabolism , Bronchi/cytology , Cytosol/metabolism , Female , Humans , Mice , Models, Biological , Mutation , Organophosphonates/metabolism , Phospholipases A2/metabolism , Pseudomonas Infections/diagnosis , Thromboplastin/metabolism
3.
Cell Microbiol ; 7(12): 1811-22, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16309466

ABSTRACT

As Pseudomonas aeruginosa ExoU possesses two functional blocks of homology to calcium-independent (iPLA(2)) and cytosolic phospholipase A(2) (cPLA(2)), we addressed the question whether it would exhibit a proinflammatory activity by enhancing the synthesis of eicosanoids by host organisms. Endothelial cells from the HMEC-1 line infected with the ExoU-producing PA103 strain exhibited a potent release of arachidonic acid (AA) that could be significantly inhibited by methyl arachidonyl fluorophosphonate (MAFP), a specific PLA(2) inhibitor, as well as significant amounts of the cyclooxygenase (COX)-derived prostaglandins PGE(2) and PGI(2). Cells infected with an isogenic mutant defective in ExoU synthesis did not differ from non-infected cells in the AA release and produced prostanoids in significantly lower concentrations. Infection by PA103 induced a marked inflammatory response in two different in vivo experimental models. Inoculation of the parental bacteria into mice footpads led to an early increase in the infected limb volume that could be significantly reduced by inhibitors of both COX and lipoxygenase (ibuprofen and NDGA respectively). In an experimental respiratory infection model, bronchoalveolar lavage (BAL) from mice instilled with 10(4) cfu of PA103 exhibited a marked influx of inflammatory cells and PGE(2) release that could be significantly reduced by indomethacin, a non-selective COX inhibitor. Our results suggest that ExoU may contribute to P. aeruginosa pathogenesis by inducing an eicosanoid-mediated inflammatory response of host organisms.


Subject(s)
Eicosanoids/biosynthesis , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arachidonic Acid/antagonists & inhibitors , Arachidonic Acid/metabolism , Arachidonic Acids/pharmacology , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cell Line , Dinoprostone/metabolism , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Epoprostenol/metabolism , Female , Group IV Phospholipases A2 , Humans , Ibuprofen/therapeutic use , Indomethacin/therapeutic use , Inflammation/pathology , Lipoxygenase Inhibitors/therapeutic use , Masoprocol/therapeutic use , Mice , Mice, Inbred BALB C , Organophosphonates/pharmacology , Phospholipases A/antagonists & inhibitors , Pseudomonas Infections/drug therapy , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/pathogenicity
4.
Int Endod J ; 38(9): 604-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16104973

ABSTRACT

AIM: To evaluate the cytotoxic effects of two brands of mineral trioxide aggregate (MTA) (Pro-Root MTA and MTA Angelus) and Portland cement (PC) on the human ECV 304 endothelial cell line. METHODOLOGY: Endothelial ECV 304 cells were incubated at 37 degrees C in an atmosphere of 95% air, 5% carbon dioxide and 100% humidity for 7 days and grown in F12 medium supplemented with 10% fetal bovine serum with 50 microg mL(-1) of gentamicin sulphate. Effects of the materials on mitochondrial functions were measured by a colorimetric assay. At each experimental time interval (24, 48 and 72 h), a dimethyl-thiazol-diphenyl tetrazolium bromid assay was conducted to measure cell viability. All assays were repeated three times to ensure reproducibility. Results were expressed as average absorbance (A(570/nm)) +/- SD and the data were analysed statistically by one-way analysis of variance and the Bonferroni post-test. A P-value < 0.05 was considered statistically significant. RESULTS: No statistically significant difference was shown between any of the experimental materials (P > 0.05). CONCLUSIONS: The two brands of MTA analysed, as well as the PC, initially showed a similar elevated cytotoxic effect that decreased gradually with time allowing the cell culture to become reestablished.


Subject(s)
Endothelial Cells/drug effects , Root Canal Filling Materials/toxicity , Aluminum Compounds/toxicity , Analysis of Variance , Calcium Compounds/toxicity , Cell Line, Transformed , Cell Survival/drug effects , Coloring Agents/metabolism , Dental Cements/toxicity , Drug Combinations , Humans , Linear Models , Mitochondria/drug effects , Mitochondria/metabolism , Oxides/toxicity , Silicates/toxicity , Tetrazolium Salts/metabolism , Thiazoles/metabolism
5.
Microbiol Immunol ; 45(8): 563-9, 2001.
Article in English | MEDLINE | ID: mdl-11592630

ABSTRACT

Bacteria of Stenotrophomonas maltophilia have been isolated with increasing frequency from the airways of cystic fibrosis (CF) patients, usually following P. aeruginosa infections, but their adherence to human epithelial respiratory cells has never been investigated. In this study, various S. maltophilia strains were seen to adhere to epithelial respiratory cells in vitro, mainly along intercellular junctions. Bacteria could also enter into host cells, as determined by the gentamicin exclusion assay and transmission electron microscopy. Cells co-incubated with P. aeruginosa and S. maltophilia exhibited a significantly decreased adherence of these latter bacteria. No decrease in S. maltophilia adherence was observed when co-infection was carried out with heat-killed P. aeruginosa or when respiratory cells were first incubated with P. aeruginosa, before incubation with S. maltophilia. Our data suggest that P. aeruginosa infections do not account for the increased prevalence of S. maltophilia in CF patient airways, that thermolabile products from P. aeruginosa can control the adherence of S. maltophilia to respiratory cells and also that these two bacteria do not compete for cell receptors.


Subject(s)
Bacterial Adhesion , Respiratory Mucosa/microbiology , Stenotrophomonas maltophilia/pathogenicity , Bronchi/cytology , Bronchi/microbiology , Cystic Fibrosis/microbiology , Epithelial Cells/cytology , Epithelial Cells/microbiology , Humans , Intercellular Junctions/microbiology , Pseudomonas aeruginosa/pathogenicity , Respiratory Mucosa/cytology
6.
Microb Pathog ; 29(6): 345-56, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11095919

ABSTRACT

Pseudomonas aeruginosa has been shown to enter into human endothelial cells in vitro. To ascertain the effects of bacterial intracellular (IC) infection, endothelial cells were exposed to PAK and PAO-1 strains for 1 h and treated with gentamicin in culture medium for different periods. P. aeruginosa induced a significant production of superoxide and hydrogen peroxide by endothelial cells. Concentrations of IC bacteria were reduced progressively with time and no viable PAO-1 was detected at 24 h after infection. However, IC infection led to killing of 32.2%+/-2.9 and 51.8%+/-3.5 of the cells infected with PAK and PAO-1, respectively, as determined by the MTT assay. By three criteria (transmission electron microscopy, DNA electrophoresis and reactivity with annexin V) infected cells exhibited features of apoptosis. Treatment of infected cells with anti-oxidants (catalase, tocopherol and N -acetyl-L-cysteine) significantly decreased the percentage of cell death. In contrast, treatment with aminoguanidine, an inhibitor of inducible NO synthase, increased significantly the killing of PAO-1 infected cells. Based on these results we speculate that in response to P. aeruginosa infection, endothelial cells increase the production of reactive oxygen intermediates to eliminate IC pathogens, but cells do not resist the oxidative stress and die by apoptosis.


Subject(s)
Apoptosis/physiology , Endothelium/microbiology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/pathogenicity , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Catalase/pharmacology , Cell Line , DNA, Bacterial/chemistry , DNA, Bacterial/isolation & purification , Electrophoresis, Agar Gel , Endothelium/cytology , Flow Cytometry , Fluorescent Dyes/chemistry , Formazans/chemistry , Guanidines/pharmacology , Humans , Lipopolysaccharides , Microscopy, Electron , Microscopy, Fluorescence , Phenanthridines/chemistry , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/genetics , Reactive Oxygen Species/physiology , Tetrazolium Salts/chemistry , Vitamin E/pharmacology
7.
Immunology ; 101(2): 271-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11012781

ABSTRACT

Proinflammatory cytokines have been shown to activate endothelial cells. To investigate the effect of cytokines on the interaction of human umbilical vein endothelial cells (HUVEC) with Pseudomonas aeruginosa, cells were treated with interferon-gamma (IFN-gamma) plus tumour necrosis factor-alpha (TNF-alpha) for 24 hr and exposed to P. aeruginosa suspension for 1 hr. Light microscopy showed that activated cells internalized significantly more bacteria than control cells. To ascertain the effect of cytokines on the microbicidal activity of HUVEC, the concentrations of viable intracellular (IC) bacteria in control and activated cells were determined, at 1 and 5 hr postinfection, by the gentamicin exclusion assay. In control cells, no significant decrease in the concentration of bacteria was detected 5 hr postinfection. In contrast, in activated cells the concentration of viable bacteria at 5 hr was significantly lower. Concentrations of superoxide and hydrogen peroxide detected in supernatants of activated cells were significantly higher than in control cell supernatants. HUVEC anti-P. aeruginosa activity was insensitive to the antioxidants superoxide dismutase, dimethylthiourea and allopurinol as well as to the L-arginine analogues aminoguanidine and NG-monomethyl-L-arginine (L-NMMA), but was significantly inhibited by catalase. Our results indicate that HUVEC can be activated by IFN-gamma plus TNF-alpha to kill IC P. aeruginosa and suggest a role for reactive oxygen radicals, notably hydrogen peroxide, in HUVEC antibacterial activity.


Subject(s)
Endothelium, Vascular/immunology , Interferon-gamma/immunology , Pseudomonas aeruginosa/immunology , Tumor Necrosis Factor-alpha/immunology , Cell Culture Techniques , Cell Survival/immunology , Endocytosis/immunology , Endothelium, Vascular/microbiology , Humans , Nitric Oxide/immunology , Reactive Oxygen Species/immunology , Umbilical Veins/immunology , Umbilical Veins/microbiology
8.
Eur Respir J ; 13(6): 1301-9, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10445605

ABSTRACT

Initial infection of the airway by Pseudomonas aeruginosa may occur through a variety of bacterial strategies including binding to epithelial receptors present at the surface of the respiratory epithelium. In order to characterize the adherence sites for P. aeruginosa in damaged and repairing bronchial tissue, an ex vivo model of airway epithelial injury and repair was developed using primary cell cultures of nasal cells from 14 subjects with polyposis. P. aeruginosa strongly adhered to flattened dedifferentiated (FD) bronchial and nasal cytokeratin 13-positive epithelial cells in the process of migration for repair. In in vitro experiments, competitive binding inhibition assays demonstrated that alpha5beta1 integrins and cellular fibronectin, in particular the RGD sequence, are receptors involved in P. aeruginosa adherence to FD nasal epithelial cells. Fluorescent cell sorting analysis and immunofluorescence techniques revealed that the alpha5beta1 integrins are overexpressed and apically exposed in FD nasal epithelial cells. One 50 kDa outer membrane protein was identified in piliated and nonpiliated strains of P. aeruginosa that was involved in binding to cellular fibronectin and alpha5beta1 epithelial integrins. These results demonstrate that Pseudomonas aeruginosa adherence is related to the dedifferentiation of airway epithelium during the repair process which unmasks and upregulates the alpha5beta1 integrin expression and induces active synthesis of cellular fibronectin. These epithelial receptors are then used by a Pseudomonas aeruginosa 50 kDa outer membrane protein as sites of bacterial adherence.


Subject(s)
Bacterial Adhesion/physiology , Bronchi/microbiology , Fibronectins/physiology , Nasal Mucosa/microbiology , Pseudomonas aeruginosa/physiology , Receptors, Fibronectin/physiology , Bacterial Outer Membrane Proteins/physiology , Bronchi/metabolism , Bronchi/pathology , Cell Differentiation , Cells, Cultured , Epithelium/metabolism , Epithelium/microbiology , Epithelium/pathology , Fibronectins/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Nasal Mucosa/metabolism , Nasal Mucosa/pathology , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Receptors, Fibronectin/metabolism , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/pathology
9.
Am J Respir Cell Mol Biol ; 20(5): 880-90, 1999 May.
Article in English | MEDLINE | ID: mdl-10226058

ABSTRACT

Internalization of Pseudomonas aeruginosa by epithelial respiratory cell lines has been suggested to be dependent on the cystic fibrosis transmembrane conductance regulator (CFTR) protein. Because we have observed intracellular (IC) P. aeruginosa only in cells that do not express apical CFTR, we addressed the question of whether bacterial internalization by epithelial cells depends on the degree of cell differentiation and polarity. Internalization of piliated P. aeruginosa PAO-1 and PAK by human epithelial respiratory cells in primary culture and by the 16 human bronchial epithelial 14o- cell line cultured either on thick collagen gels or on thin collagen films was evaluated by the gentamicin exclusion assay. Cells cultured on thick gels were differentiated, polarized, and tight. They exhibited CFTR at their apical membranes, expressed beta1 integrins at their basal membranes, excluded lanthanum nitrate, and uniformly expressed ZO-1 protein. In contrast, in cells cultured on thin films, CFTR was present mainly in the cytoplasm, whereas beta1 integrins were detected at apical membranes. Most cells cultured on thin films did not exclude lanthanum nitrate and rarely expressed ZO-1 protein. Cells grown on thick and thin collagen substrates differed markedly in bacterial internalization: no IC bacteria could be detected in cells cultured on gels, whereas high IC bacterial concentrations were isolated from cells cultured on thin films. Treatment of cells cultured on thin films with ethylenediaminetetraacetic acid, to disrupt intercellular junctions further, significantly enhanced P. aeruginosa internalization. Our results suggest that P. aeruginosa internalization by epithelial respiratory cells does not depend on CFTR protein expression at the epithelial cell surface but rather on cell polarity and junctional complex integrity.


Subject(s)
Bronchi/microbiology , Pseudomonas aeruginosa/physiology , Bronchi/cytology , Bronchi/ultrastructure , Cell Differentiation , Cell Polarity , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/microbiology , Epithelial Cells/ultrastructure , Fluorescent Antibody Technique , Humans , Microscopy, Electron
10.
J Leukoc Biol ; 65(4): 508-14, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10204580

ABSTRACT

Intravenous administration of lipopolysaccharide (LPS) to rats increased the production of nitric oxide (NO) metabolites (NOx) by blood polymorphonuclear neutrophils (PMN) in vitro. Both dexamethasone and L-NMMA, added in vitro to neutrophil cultures, inhibited the production of NO. On the other hand, the production of NO was not affected by the treatment, in vivo or in vitro, with different inhibitors of cyclooxygenase or 5-lipoxygenase or with a platelet-activating factor (PAF) antagonist. The incubation of blood PMN from normal rats in vitro with neutrophil activators (PAF, leukotriene B4, and interleukin-8) and different cytokines [interleukin-1, tumor necrosis factor alpha, and interferon-gamma (IFN-gamma)] showed that only IFN-gamma was able to induce the production of high amounts of NO. This induction was directly correlated with the expression of iNOS and an increase in in the enzyme activity in blood PMN. The tyrosine kinase inhibitor genistein inhibited NO production induced by IFN-gamma, suggesting that the signal transduction pathway leading to NOS induction in rat PMN involves phosphorylation by tyrosine kinase. We also showed that NO produced by IFN-gamma activated rat blood PMN involved in the killing of Pseudomonas aeruginosa.


Subject(s)
Blood Bactericidal Activity/immunology , Neutrophils/immunology , Nitric Oxide Synthase/immunology , Protein-Tyrosine Kinases/biosynthesis , Animals , Blood Bactericidal Activity/drug effects , Cytokines/physiology , Dexamethasone/pharmacology , Enzyme Induction/drug effects , Enzyme Induction/immunology , Interferon-gamma/pharmacology , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Male , Neutrophils/drug effects , Neutrophils/enzymology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/blood , Nitric Oxide Synthase Type II , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/immunology , Rats , Rats, Wistar , omega-N-Methylarginine/pharmacology
11.
Biocell ; 23(1): 65-72, Apr. 1999.
Article in English | LILACS | ID: lil-340374

ABSTRACT

Release of lactate dehydrogenase (LDH) from the cytoplasmic compartment, trypan blue exclusion and methylthiazole tetrazolium (MTT) colorimetric assays were compared with regard to their sensitivity in detecting damage of human cultured epithelial cells induced by sodium fluoride or puromycin. LDH assay did not detect any difference between controls and cells treated with either of the two drugs. Cell monolayers treated with 0.3 sodium fluoride or 10(-2) M puromycin presented higher percentages of cells that took up the trypan blue dye than controls but monolayers treated with lower drug concentrations did not differ from controls. Viability measured by MTT assay was the most sensitive assay, detecting a dose-dependent impairment of cell function after treatment with the two drugs. Moreover, MTT offered major advantages in speed, simplicity and precise quantitation over the other viability assays


Subject(s)
Humans , Animals , Cell Culture Techniques , Liver/cytology , Cell Survival , Chlorocebus aethiops , Coloring Agents , L-Lactate Dehydrogenase , Mammals , Tetrazolium Salts , Thiazoles , Trypan Blue , Vero Cells
12.
Biocell ; 23(1): 65-72, Apr. 1999.
Article in English | BINACIS | ID: bin-6060

ABSTRACT

Release of lactate dehydrogenase (LDH) from the cytoplasmic compartment, trypan blue exclusion and methylthiazole tetrazolium (MTT) colorimetric assays were compared with regard to their sensitivity in detecting damage of human cultured epithelial cells induced by sodium fluoride or puromycin. LDH assay did not detect any difference between controls and cells treated with either of the two drugs. Cell monolayers treated with 0.3 sodium fluoride or 10(-2) M puromycin presented higher percentages of cells that took up the trypan blue dye than controls but monolayers treated with lower drug concentrations did not differ from controls. Viability measured by MTT assay was the most sensitive assay, detecting a dose-dependent impairment of cell function after treatment with the two drugs. Moreover, MTT offered major advantages in speed, simplicity and precise quantitation over the other viability assays


Subject(s)
Humans , Animals , Comparative Study , Cell Culture Techniques/methods , Liver/cytology , Chlorocebus aethiops , Cell Survival , Coloring Agents , L-Lactate Dehydrogenase/metabolism , Mammals , Tetrazolium Salts , Thiazoles , Trypan Blue , Vero Cells
13.
Biocell ; 23(1): 65-72, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10904534

ABSTRACT

Release of lactate dehydrogenase (LDH) from the cytoplasmic compartment, trypan blue exclusion and methylthiazole tetrazolium (MTT) colorimetric assays were compared with regard to their sensitivity in detecting damage of human cultured epithelial cells induced by sodium fluoride or puromycin. LDH assay did not detect any difference between controls and cells treated with either of the two drugs. Cell monolayers treated with 0.3% sodium fluoride or 10(-2) M puromycin presented higher percentages of cells that took up the trypan blue dye than controls but monolayers treated with lower drug concentrations did not differ from controls. Viability measured by MTT assay was the most sensitive assay, detecting a dose-dependent impairment of cell function after treatment with the two drugs. Moreover, MTT offered major advantages in speed, simplicity and precise quantitation over the other viability assays.


Subject(s)
Cell Culture Techniques/methods , Cell Survival , Liver/cytology , Animals , Chlorocebus aethiops , Coloring Agents , Humans , L-Lactate Dehydrogenase/metabolism , Mammals , Tetrazolium Salts , Thiazoles , Trypan Blue , Vero Cells
14.
Biocell ; 23(1): 65-72, 1999 Apr.
Article in English | BINACIS | ID: bin-40091

ABSTRACT

Release of lactate dehydrogenase (LDH) from the cytoplasmic compartment, trypan blue exclusion and methylthiazole tetrazolium (MTT) colorimetric assays were compared with regard to their sensitivity in detecting damage of human cultured epithelial cells induced by sodium fluoride or puromycin. LDH assay did not detect any difference between controls and cells treated with either of the two drugs. Cell monolayers treated with 0.3


sodium fluoride or 10(-2) M puromycin presented higher percentages of cells that took up the trypan blue dye than controls but monolayers treated with lower drug concentrations did not differ from controls. Viability measured by MTT assay was the most sensitive assay, detecting a dose-dependent impairment of cell function after treatment with the two drugs. Moreover, MTT offered major advantages in speed, simplicity and precise quantitation over the other viability assays.

15.
Microb Pathog ; 23(4): 249-55, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9344786

ABSTRACT

Human respiratory cells participating in the repair of epithelial wounds have been shown to be highly susceptible to Pseudomonas aeruginosa adherence. To ascertain whether such susceptibility is a common feature of different repairing epithelial cells, Caco-2 cell monolayers were chemically injured, reincubated for 48 h to partially repair and exposed to bacteria. Cells edging the wounds that spread and migrate to re-establish cell confluence were called 'repairing cells' while cells far from the wounds were called 'non-repairing cells'. By light microscopy, bacteria were seen to adhere to and to enter into both repairing and non-repairing cells. The percentage of intracellular bacteria in repairing cells was significantly higher than in non-repairing cells. The higher susceptibility of repairing monolayers to bacterial entry was confirmed by the gentamicin exclusion assay. P. aeruginosa entry into Caco-2 cells was greatly enhanced in non-injured confluent monolayers treated with EDTA to disrupt intercellular junctions. As tight junction disfunctions have been described during the wound repair process, we speculate that exposure of basolateral receptors to bacterial ligands may account for the enhancement of P. aeruginosa internalization by repairing monolayers.


Subject(s)
Epithelial Cells/microbiology , Pseudomonas aeruginosa/growth & development , Wound Healing , Bacterial Adhesion , Caco-2 Cells , Cell Polarity , Epithelial Cells/pathology , Humans , Opportunistic Infections/etiology , Pseudomonas Infections/etiology , Pseudomonas aeruginosa/pathogenicity , Receptors, Cell Surface
16.
J Submicrosc Cytol Pathol ; 29(2): 229-37, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9165717

ABSTRACT

P. aeruginosa is selectively internalized by human endothelial cells but is not efficiently killed in the intracellular (IC) compartment. To investigate whether IC survival is associated with failure in bacteria-containing endosome-lysosome (E-L) fusion, endothelial cells were exposed to albumin-colloidal gold complex and to bacterial suspension and submitted to transmission electron microscopy (TEM). Gold granules were detected in P. aeruginosa-containing vacuoles, indicating that E-L fusion had occurred. Bacteria were also seen apparently free in the cell cytoplasm, suggesting disruption of endosome membranes. To ascertain whether phospholipase C (PLC) could account for vacuolar lysis, PLC producing PAO1 and PAK strains were compared with a PLC deficient mutant (PLCN) in their IC survival. All three strains were equally uptaken by the endothelial cells, as determined by the gentamicin exclusion assay. After 3 h of infection, the IC concentration of PAK and PAO1 increased significantly while the concentration of the mutant decreased to 56.8 +/- 18.2% of the viable counts at 1 h of infection. After 5 h, the IC concentration of P. aeruginosa corresponded to 83.1 +/- 34.6%, 109 +/- 22.6% and 26.2 +/- 14.7% of the viable counts detected at 1 h, for PAK, PAO1 and the mutant, respectively. By TEM, while most PAO1-containing vacuoles presented partially lysed membranes, in cells infected with the PLCN mutant bacteria were most often observed in vacuoles with intact membranes. These observations suggest that the IC survival of P. aeruginosa results from a competition between the microbicidal activity of endothelial cells following E-L fusion and the capacity of bacteria to escape from endosomes.


Subject(s)
Endosomes/ultrastructure , Endothelium, Vascular/microbiology , Endothelium, Vascular/ultrastructure , Phagosomes/ultrastructure , Pseudomonas aeruginosa/physiology , Cell Survival , Cells, Cultured , Chloroquine/pharmacology , Cytoplasm/microbiology , Cytoplasm/ultrastructure , Endosomes/physiology , Endothelium, Vascular/physiology , Humans , Intracellular Membranes/physiology , Intracellular Membranes/ultrastructure , Membrane Fusion , Microscopy, Electron , Phagosomes/physiology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/ultrastructure , Type C Phospholipases/metabolism , Vacuoles/microbiology , Vacuoles/physiology , Vacuoles/ultrastructure
17.
Infect Immun ; 64(5): 1582-8, 1996 May.
Article in English | MEDLINE | ID: mdl-8613364

ABSTRACT

We investigated the implication of asialo GM1 as an epithelial receptor in the increased Pseudomonas aeruginosa affinity for regenerating respiratory epithelial cells from cystic fibrosis (CF) and non-CF patients. Human respiratory epithelial cells were obtained from nasal polyps of non-CF subjects and of CF patients homozygous for the delta F 508 transmembrane conductance regulator protein (CFTR) mutation and cultured according to the explant-outgrowth model. At the periphery of the outgrowth, regenerating respiratory epithelial cells spreading over the collagen I matrix with lamellipodia were observed, characteristic of respiratory epithelial wound repair after injury. P aeruginosa adherence to regenerating respiratory epithelial cells was found to be significantly greater in the delta F 508 homozygous CF group than in the non-CF group (P < 0.001). In vitro competitive binding inhibition assays performed with rabbit polyclonal antibody against asialo GM1 demonstrated that blocking asialo GM1 reduces P. aeruginosa adherence to regenerating respiratory epithelial cells in delta F 508 homozygous cultures (P < 0.001) as well as in non-CF cultures (P < 0.001). Blocking of asialo GM1 was significantly more efficient in CF patients than in non-CF subjects (P < 0.05). Distribution of asialo GM1 as determined by preembedding labelling and immunoelectron microscopy clearly demonstrated the specific apical membrane expression of asialo GM1 by regenerating respiratory epithelial cells, whereas other cell phenotypes did not apically express asialo GM1. These results demonstrate that (i) asialo GM1 is an apical membrane receptor for P. aeruginosa expressed at the surface of CF and non-CF regenerating respiratory epithelial cells and (ii) asialo GM1 is specifically recovered in regenerating respiratory epithelium. These results suggest that in CF, epithelial repair represents the major event which exposes asialo GM1 for P. aeruginosa adherence.


Subject(s)
Bacterial Adhesion/physiology , G(M1) Ganglioside/physiology , Pseudomonas aeruginosa/pathogenicity , Respiratory System/microbiology , Adolescent , Adult , Animals , Cells, Cultured , Child , Cystic Fibrosis/complications , Cystic Fibrosis/genetics , Cystic Fibrosis/microbiology , Epithelium/microbiology , Epithelium/physiology , Epithelium/ultrastructure , Female , G(M1) Ganglioside/immunology , Humans , Male , Microscopy, Electron, Scanning , Microscopy, Immunoelectron , Opportunistic Infections/etiology , Opportunistic Infections/microbiology , Pseudomonas Infections/etiology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/physiology , Rabbits , Regeneration , Respiratory Physiological Phenomena , Respiratory System/ultrastructure , Respiratory Tract Infections/etiology , Respiratory Tract Infections/microbiology
18.
Infect Immun ; 64(2): 600-5, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8550213

ABSTRACT

Pseudomonas aeruginosa is a major human pathogen known to infect tissues that have been previously damaged in some way. In wounded human respiratory tissues, P. aeruginosa cells were found attached to exposed basement membranes following epithelial denudation, suggesting that the affinity for extracellular matrix proteins may account for the bacterium's opportunistic character. By using microtiter wells coated with different P. aeruginosa strains, we demonstrated that laminin binds to both colonizing bacterial strains, isolated from asymptomatic carriers, and strains isolated from infected patients. Binding of soluble laminin to piliated P. aeruginosa PAK and to the nonpiliated isogenic mutant PAK/p--was shown to be saturable. Binding of laminin to the piliated PAK strain was not different from binding to th nonpiliated PAK/p--strain but was significantly higher than binding to the avirulent, nonpiliated PAK-N1 rpoN mutant. By transmission electron microscopy, we localized the laminin-binding sites on a loose material in the outermost layer of the bacteria. Western immunoblotting results suggested that 57- and 59-kDa nonpilus adhesins from the microbial outer membranes account for the binding of P. aeruginosa to laminin. We speculate that bacterial affinity for laminin may be of biological significance in the pathogenesis of P. aeruginosa infection of injured tissues.


Subject(s)
Adhesins, Bacterial/analysis , Bacterial Adhesion , Laminin/metabolism , Lectins , Pseudomonas aeruginosa/physiology , Adhesins, Bacterial/metabolism , Binding Sites , Humans
19.
Infect Immun ; 62(12): 5456-63, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7960126

ABSTRACT

The pathogenesis of Pseudomonas aeruginosa disseminated infections depends on bacterial interaction with blood vessels. We have hypothesized that in order to traverse the endothelial barrier, bacteria would have to adhere to and damage endothelial cells. To test this hypothesis, we studied the adherence to human endothelial cells in primary culture of the piliated P. aeruginosa strain PAK and of two isogenic nonpiliated strains: PAK/p-, which carries a mutation in the pilin structural gene, and PAK-N1, a mutant defective in the regulatory rpoN gene. PAK adhered significantly more than did the pilus-lacking strains. P. aeruginosa was also taken up by endothelial cells, as determined by quantitative bacteriologic assays and by transmission electron microscopy. This internalization of P. aeruginosa seems to be a selective process, since the piliated strain was taken up significantly more than the nonpiliated bacteria and the avirulent Escherichia coli DH5 alpha, even following bacterial centrifugation onto the cell monolayers. A significant fraction of the internalized P. aeruginosa PAK was recovered in a viable form after 6 h of residence within endothelial cells. Progressive endothelial cell damage resulted from PAK intracellular harboring, as indicated by the release of lactate dehydrogenase. An increasing concentration of PAK cells was recovered from the extracellular medium with time, suggesting that ingested bacteria were released from endothelial cells and multiplied freely. We speculate that in vivo the ability of some P. aeruginosa strains to resist intracellular residence would afford protection from host defenses and antibiotics and that the release of viable bacteria into bloodstream may represent a central feature of the pathogenesis of bacteremia in compromised patients.


Subject(s)
Bacterial Adhesion/physiology , DNA-Binding Proteins , DNA-Directed RNA Polymerases , Endothelium, Vascular/microbiology , Fimbriae, Bacterial/physiology , Pseudomonas aeruginosa/physiology , Bacterial Adhesion/genetics , Bacterial Outer Membrane Proteins/genetics , Biological Transport/drug effects , Cell Division , Cells, Cultured , Cytochalasin D/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Endothelium, Vascular/ultrastructure , Escherichia coli Proteins , Fimbriae Proteins , Fimbriae, Bacterial/genetics , Genes, Bacterial/genetics , Humans , L-Lactate Dehydrogenase/analysis , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/ultrastructure , RNA Polymerase Sigma 54 , Sigma Factor/genetics , Umbilical Veins/cytology
20.
Infect Immun ; 62(10): 4481-7, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7927712

ABSTRACT

Pseudomonas aeruginosa is the most important bacterial pathogen associated with chronic airway infection, especially in cystic fibrosis. We addressed the question of whether the galactophilic internal lectin of P. aeruginosa (PA-I) could represent a virulence factor for the respiratory epithelium. PA-I lectin was localized in all the bacteria of P. aeruginosa ATCC 33347 as determined by immunofluorescence staining. We investigated the dose-dependent effect of P. aeruginosa PA-I lectin on the growth, the ciliary beating frequency, and the morphology of human respiratory cells in primary culture of nasal polyps collected from non-cystic fibrosis patients. PA-I lectin significantly (P < 0.01) inhibited the growth of respiratory cells at a concentration of > or = 10 micrograms/ml. The percentage of active ciliated cell surface of the cultures decreased significantly (P < 0.05) at a PA-I lectin concentration of 50 micrograms/ml. Exposed to a low concentration of PA-I lectin (10 micrograms/ml), respiratory epithelial cells showed intracytoplasmic vacuoles when examined by light and transmission electron microscopy. At a higher concentration of PA-I lectin (100 micrograms/ml), major cell damage and severe epithelial shedding occurred. These results demonstrate that the P. aeruginosa internal PA-I lectin has a dose-dependent cytotoxic effect on respiratory epithelial cells in vitro. The P. aeruginosa PA-I lectin may represent a virulence factor by contributing to the respiratory epithelial damage during P. aeruginosa respiratory infections.


Subject(s)
Adhesins, Bacterial , Bacterial Adhesion , Bacterial Proteins/toxicity , Lectins/toxicity , Pseudomonas aeruginosa/pathogenicity , Respiratory System/drug effects , Animals , Cells, Cultured , Epithelium/drug effects , Epithelium/pathology , Humans , Lectins/isolation & purification , Rabbits , Respiratory System/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...