Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Microbiol ; 3(11): 763-72, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11696036

ABSTRACT

The invasion of Pseudomonas aeruginosa and Salmonella enterica serovar Typhi into epithelial cells depends on the cystic fibrosis transmembrane conductance regulator (CFTR) protein as an epithelial receptor. In the case of P. aeruginosa, the bacterial ligand for CFTR is the outer core oligosaccharide portion of the lipopolysaccharide (LPS). To determine whether serovar Typhi LPS is also a bacterial ligand mediating internalization, we used both P. aeruginosa and serovar Typhi LPS as a competitive inhibitor of serovar Typhi invasion into the epithelial cell line T84. P. aeruginosa LPS containing a complete core efficiently inhibited serovar Typhi invasion. However, neither killed wild-type Typhi cells nor purified LPS were effective inhibitors. LPS from mutant Typhi strains defective in O side-chain synthesis, but with an apparently normal core, was capable of inhibiting invasion, but LPS obtained from a deeper rough mutant strain with alterations in fast-migrating core oligosaccharide failed to inhibit invasion. Lastly, exposure of wild-type serovar Typhi to T84 cultures before heat killing resulted in a structural alteration in its LPS that allowed the heat-killed cells to inhibit invasion of wild-type serovar Typhi. These data indicate that the serovar Typhi LPS core, like the P. aeruginosa LPS core, is a ligand mediating internalization of bacteria by epithelial cells, and that exposure of this ligand on wild-type Typhi is induced by the bacteria's interaction with host cells.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/microbiology , Lipopolysaccharides/chemistry , Lipopolysaccharides/pharmacology , Salmonella typhi/pathogenicity , Binding Sites , Cell Line , Humans , Ligands , Lipopolysaccharides/isolation & purification , Lipopolysaccharides/metabolism , Mutation , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/physiology , Salmonella typhi/drug effects , Salmonella typhi/physiology
2.
Infect Immun ; 67(3): 1481-92, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10024598

ABSTRACT

Previous findings indicate that the cystic fibrosis transmembrane conductance regulator (CFTR) is a ligand for Pseudomonas aeruginosa ingestion into respiratory epithelial cells. In experimental murine keratitis, P. aeruginosa enters corneal epithelial cells. We determined the importance of CFTR-mediated uptake of P. aeruginosa by corneal cells in experimental eye infections. Entry of noncytotoxic (exoU) P. aeruginosa into human and rabbit corneal cell cultures was inhibited with monoclonal antibodies and peptides specific to CFTR amino acids 108 to 117. Immunofluorescence microscopy and flow cytometry demonstrated CFTR in the intact murine corneal epithelium, and electron microscopy showed that CFTR binds to P. aeruginosa following corneal cell ingestion. In experimental murine eye infections, multiple additions of 5 nM CFTR peptide 103-117 to inocula of either cytotoxic (exoU+) or noncytotoxic P. aeruginosa resulted in large reductions in bacteria in the eye and markedly lessened eye pathology. Compared with wild-type C57BL/6 mice, heterozygous DeltaF508 Cftr mice infected with P. aeruginosa had an approximately 10-fold reduction in bacterial levels in the eye and consequent reductions in eye pathology. Homozygous DeltaF508 Cftr mice were nearly completely resistant to P. aeruginosa corneal infection. CFTR-mediated internalization of P. aeruginosa by buried corneal epithelial cells is critical to the pathogenesis of experimental eye infection, while in the lung, P. aeruginosa uptake by surface epithelial cells enhances P. aeruginosa clearance from this tissue.


Subject(s)
Cornea/microbiology , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Keratitis/etiology , Pseudomonas Infections/etiology , Animals , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Cells/microbiology , Flow Cytometry , Humans , Keratitis/pathology , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , Rabbits
3.
Proc Natl Acad Sci U S A ; 94(22): 12088-93, 1997 Oct 28.
Article in English | MEDLINE | ID: mdl-9342367

ABSTRACT

The cystic fibrosis transmembrane conductance regulator (CFTR) is a chloride ion channel, but its relationship to the primary clinical manifestation of CF, chronic Pseudomonas aeruginosa pulmonary infection, is unclear. We report that CFTR is a cellular receptor for binding, endocytosing, and clearing P. aeruginosa from the normal lung. Murine cells expressing recombinant human wild-type CFTR ingested 30-100 times as many P. aeruginosa as cells lacking CFTR or expressing mutant DeltaF508 CFTR protein. Purified CFTR inhibited ingestion of P. aeruginosa by human airway epithelial cells. The first extracellular domain of CFTR specifically bound to P. aeruginosa and a synthetic peptide of this region inhibited P. aeruginosa internalization in vivo, leading to increased bacterial lung burdens. CFTR clears P. aeruginosa from the lung, indicating a direct connection between mutations in CFTR and the clinical consequences of CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Endocytosis , Lung Diseases/microbiology , Pseudomonas Infections/microbiology , Receptors, Cell Surface/metabolism , Animals , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Endocytosis/drug effects , Epithelial Cells/microbiology , Humans , Lung/cytology , Mice , Peptide Fragments/pharmacology , Protein Binding , Receptors, Cell Surface/genetics
4.
Infect Immun ; 65(4): 1370-6, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9119476

ABSTRACT

The prevention of bacterial infections by the inhibition of binding to host tissues is an oft-touted approach, but few studies with appropriate models of infection have tested its feasibility. Pseudomonas aeruginosa causes severe corneal infections in mice after inoculations with low doses, and infection is thought to depend upon an initial adherence of the bacteria to corneal cells. In vitro, adherence to corneal cells is mediated to a large degree by the complete-outer-core oligosaccharide of the bacterial lipopolysaccharide (LPS). However, bacteria adhering to tissues in vivo are difficult to differentiate from nonadherent bacteria. Since a direct correlate of P. aeruginosa adherence to corneal epithelial cells is the degree to which these cells internalize P. aeruginosa, the level of adherence in vivo can be approximated by measuring P. aeruginosa ingestion by cells by using gentamicin exclusion assays. To determine the degree to which inhibition of the corneal cell adherence affects the course of infection and disease in the murine model, we evaluated the ability of LPS-outer-core oligosaccharide to inhibit bacterial association and entry into corneal cells and to modulate the development of disease. Mice were anesthetized, and their corneas were scratched and inoculated with virulent P. aeruginosa 6294 or PAO1, along with either 50 microg of oligosaccharide derived from LPS from P. aeruginosa PAC557 (complete outer core but no O side chains) or oligosaccharide derived from LPS of P. aeruginosa PAC1RalgC::tet (incomplete-core oligosaccharide). After 4 h, there were no differences between groups in the counts of infecting and internalized bacteria. At 24 h, the complete-core oligosaccharide decreased the levels of bacteria per eye by 70 to 99.7% compared with the levels achieved by including the incomplete-core oligosaccharide in the infectious inoculum. Epithelial cell ingestion of bacteria was comparably affected. However, the effect on disease was modest and only evident at lower challenge doses that elicited mild disease in controls and when the bacterial association and ingestion were inhibited by >99%. Overall, it appears that in the murine model of P. aeruginosa corneal infection at challenge doses of bacteria 10-fold or greater than the minimal amount needed to cause disease, the absolute level of inhibition of bacterial adherence is insufficient to reduce the bacterial counts below that which elicits disease.


Subject(s)
Bacterial Adhesion , Corneal Diseases/microbiology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa , Animals , Disease Models, Animal , Mice
5.
Am J Respir Crit Care Med ; 154(4 Pt 2): S175-82, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8876538

ABSTRACT

Patients with cystic fibrosis (CF) have a pronounced hypersusceptibility (80 to 90%) to Pseudomonas aeruginosa infection. We hypothesized that airway epithelial cell ingestion of bacteria followed by cellular desquamation may protect the lung from infection, and epithelial cells expressing mutant forms of the cystic fibrosis transmembrane conductance regulator (CFTR) may be defective in this function. We found that transformed human airway epithelial cells homozygous for the delta F508 allele of CFTR were significantly defective in uptake of P. aeruginosa compared with the same cell line complemented with the wild-type allele of CFTR. Partial membrane expression of the delta F508 CFTR protein occurs in cells grown at 26 degrees C, and under these conditions uptake of P. aeruginosa occurred at levels comparable to cells with a wild-type allele of CFTR. Epithelial cell ingestion assays using isogenic bacterial strains differing in lipopolysaccharide (LPS) phenotype, along with inhibition studies, identified the LPS-core oligosaccharide as the bacterial ligand for epithelial cell invasion. Inhibition of epithelial cell ingestion of P. aeruginosa in a neonatal mouse lung infection model led to increased levels of bacteria in the lungs 24 and 48 h after infection. Defective epithelial cell internalization of P. aeruginosa may be a critical factor in hypersusceptibility of CF patients to chronic lung infections.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis/microbiology , Point Mutation , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/physiology , Respiratory Tract Infections/microbiology , Animals , Bacterial Adhesion , Cell Line, Transformed , Cystic Fibrosis/complications , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Disease Susceptibility , Epithelium/metabolism , Epithelium/microbiology , Humans , Mice , Pseudomonas Infections/complications , Pseudomonas aeruginosa/genetics , Respiratory System/metabolism , Respiratory System/microbiology , Respiratory Tract Infections/complications
6.
Optom Vis Sci ; 73(9): 590-4, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8887402

ABSTRACT

BACKGROUND: Staphylococcus epidermidis is a major causative agent of infectious keratitis associated with contact lens wear. Adhesion of this bacterium to contact lenses may contribute to the pathogenesis of infection and could be influenced by lens surface properties, packaging/storage solutions, and vary among different strains according to the level or type of adhesins expressed. METHODS: Adhesion of six clinical isolates of S. epidermidis to three different contact lens materials was tested. Adhesion assays were performed on lenses immediately after removal from their packages, and also after lenses were soaked in sterile phosphate buffered saline (PBS) for 7 days to dilute the packaging solution. RESULTS: For lenses tested immediately upon removal from their packaging, adhesion to polymacon (in PBS with 0.1% polyvinyl alcohol) was significantly greater than to etafilcon A (in borate buffered saline) and vifilcon A (in PBS). After soaking, adhesion to polymacon lenses was significantly less than to the other lens materials. This pattern was consistent for all strains, although major differences in baseline adhesion levels existed between strains, with exopolysaccharide (slime)-positive bacteria being more adherent to lenses. CONCLUSIONS: Properties of contact lens materials were not the sole determinant of viable S. epidermidis adhesion to lenses. Strain variability, including levels of exopolysaccharide expression, and the solution used for lens immersion also influenced adhesion.


Subject(s)
Bacterial Adhesion/physiology , Contact Lenses, Hydrophilic , Staphylococcus epidermidis/physiology , Humans , Staphylococcus epidermidis/isolation & purification
7.
Infect Immun ; 64(6): 2288-94, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8675339

ABSTRACT

We have reported that some strains of Pseudomonas aeruginosa can enter corneal epithelial cells during experimental murine eye infection and when the cells are cultured in vitro. Following invasion, both the host cell and the intracellular bacteria can remain viable for up to 24 h. Others have reported that toxin-mediated damage of epithelial cells contributes to the pathogenesis of P. aeruginosa keratitis. To clarify the relationship between cell invasion and cytotoxicity, fourteen P. aeruginosa isolates were compared for their capacity to enter epithelial cells and for their ability to induce cytotoxicity. Bacterial invasion was quantified by gentamicin survival assays both in vivo and in vitro. Cytotoxicity was examined qualitatively by trypan blue exclusion assays and quantitatively by chromium release assays in vitro. A significant inverse correlation was found between the ability to induce cytotoxicity and epithelial cell invasion as measured by gentamicin survival assays. Both cytotoxic and noncytotoxic strains were identified among corneal and noncorneal isolates; all isolates that were not cytotoxic were capable of epithelial cell invasion. Efficient host cell invasion could not be demonstrated for cytotoxic strains; however, the gentamicin survival assay relies upon host cells retaining viability in order to yield useful results, and this may limit the effectiveness of this assay for testing epithelial cell invasion by cytotoxic strains. Since all of the corneal isolates that were tested were virulent in vivo, the results show that there are at least two different types of P. aeruginosa-induced disease, one caused by strains that are cytotoxic and the other involving bacteria that can enter epithelial cells and survive intracellularly without killing the host cell.


Subject(s)
Cornea/microbiology , Pseudomonas aeruginosa/pathogenicity , Animals , Cells, Cultured , Cornea/pathology , Epithelium/microbiology , Gentamicins/pharmacology , Humans , Mice , Mice, Inbred C57BL , Microscopy, Electron , Rabbits
8.
Invest Ophthalmol Vis Sci ; 37(6): 976-86, 1996 May.
Article in English | MEDLINE | ID: mdl-8631641

ABSTRACT

PURPOSE: Pseudomonas aeruginosa has been observed to be adherent to and inside epithelial cells during experimental corneal infection. The authors identified bacterial ligands involved in adherence and entry of P. aeruginosa into corneal epithelial cells. METHODS: In vitro gentamicin survival assays were used to determine the intracellular survival of a panel of P. aeruginosa mutants. Strains (10(6) to 10(7) colony-forming units) were added to primary cultures of rabbit corneal epithelial cells (approximately 10(5)/well) for 3 hours, nonadherent bacteria were washed away, and extracellular bacteria were killed with gentamicin. The antibiotic was then washed away, and epithelial cells were lysed with 0.5% Triton X-100 to release internalized bacteria. Bacterial association (sum of bound and internalized bacteria) was measured by the omission of gentamicin. Similar assays were carried out with whole mouse eyes in situ. RESULTS: A lipopolysaccharide core with an exposed terminal glucose residue was found to be necessary for maximal association and entry of P. aeruginosa into corneal cells. Bacterial pili and flagella were not involved. Mutants of P. aeruginosa strains that do not produce an LPS core with a terminal glucose residue had a significantly lower level of association with (approximately 50%) and ingestion by ( > 90%, P < 0.01) corneal cells than did strains with this characteristic. Complementation of the LPS productions defect by plasmid-borne DNA returned association and ingestion to near parental levels. Lipopolysaccharides and delipidated oligosaccharides with a terminal glucose residue in the core inhibited bacterial association and entry into corneal cells. Experiments using P. aeruginosa LPS mutants and corneal cells on whole mouse eyes confirmed the role of the LPS core in cellular entry. CONCLUSIONS: Corneal epithelial cells bind and internalized P. aeruginosa by the exposed LPS core.


Subject(s)
Bacterial Adhesion , Cornea/metabolism , Cornea/microbiology , Lipopolysaccharides/metabolism , Phagocytosis/physiology , Pseudomonas aeruginosa/physiology , Animals , Anti-Bacterial Agents/pharmacology , Carbohydrate Sequence , Cells, Cultured , Chromosomes, Bacterial , Colony Count, Microbial , Cornea/cytology , Corneal Ulcer/microbiology , Epithelial Cells , Epithelium/metabolism , Epithelium/microbiology , Gentamicins/pharmacology , Ligands , Lipopolysaccharides/chemistry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oligosaccharides/metabolism , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Rabbits , Virulence
9.
Science ; 271(5245): 64-7, 1996 Jan 05.
Article in English | MEDLINE | ID: mdl-8539601

ABSTRACT

Cystic fibrosis (CF) patients are hypersusceptible to chronic Pseudomonas aeruginosa lung infections. Cultured human airway epithelial cells expressing the delta F508 allele of the cystic fibrosis transmembrane conductance regulator (CFTR) were defective in uptake of P. aeruginosa compared with cells expressing the wild-type allele. Pseudomonas aeruginosa lipopolysaccharide (LPS)-core oligosaccharide was identified as the bacterial ligand for epithelial cell ingestion; exogenous oligosaccharide inhibited bacterial ingestion in a neonatal mouse model, resulting in increased amounts of bacteria in the lungs. CFTR may contribute to a host-defense mechanism that is important for clearance of P. aeruginosa from the respiratory tract.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Cystic Fibrosis/complications , Pseudomonas Infections/etiology , Pseudomonas aeruginosa/physiology , Respiratory System/microbiology , Respiratory Tract Infections/etiology , Animals , Animals, Newborn , Cell Line, Transformed , Cystic Fibrosis/genetics , Cystic Fibrosis/microbiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Susceptibility , Epithelium/microbiology , Humans , Lipopolysaccharides/pharmacology , Lung/microbiology , Mice , Mice, Inbred BALB C , Pseudomonas Infections/microbiology , Respiratory Tract Infections/microbiology
10.
Infect Immun ; 63(10): 4072-7, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7558321

ABSTRACT

Pseudomonas aeruginosa is usually considered an extracellular pathogen. Using assays to determine intracellular survival in the presence of gentamicin, we have previously demonstrated that P. aeruginosa is able to invade corneal cells during infectious keratitis in mice. In vitro, P. aeruginosa was found to enter the following cells: human corneal cells removed by irrigation; epithelial cells in the cornea of rats, mice, and rabbits; and primary corneal epithelial cells cultured from rat and rabbit eyes. The level of invasion was related to the level of adherent or associated bacteria. In general, invasion was more efficient with cultured epithelial cells than with cells tested in situ. Invasion did not occur when assays were performed at 4 degrees C. Cytochalasin D but not colchicine inhibited bacterial invasion, suggesting that bacterial entry was an endocytic process dependent on actin microfilaments but not microtubules. Bacteria that invaded cultured corneal epithelial cells were found to multiply within cells. The ability of P. aeruginosa to invade and multiply within corneal epithelial cells may contribute to the virulence of this organism during infectious keratitis, since intracellular bacteria can evade host immune effectors and antibiotics commonly used to treat infection.


Subject(s)
Cornea/microbiology , Pseudomonas aeruginosa/physiology , Animals , Cytochalasin D/pharmacology , Epithelium/microbiology , Gentamicins/pharmacology , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Temperature
11.
Infect Immun ; 63(9): 3497-501, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7642283

ABSTRACT

A murine corneal scratch model has been used extensively to study various aspects of the pathogenesis of Pseudomonas aeruginosa, a common etiologic agent of corneal infections. This model uses mild inhalation anesthetics which keep the animals immobile for a relatively short time and promote the interaction between the infecting organisms and the corneal wound. Under these circumstances, only a small number of P. aeruginosa isolates delivered at inocula of > 10(7) CFU are infectious. We determined that this model is useful for studying other P. aeruginosa strains given at lower doses if injectable anesthetics are administered prior to infection to keep the animals immobile for 15 to 30 min. Under these conditions, eight clinical isolates of P. aeruginosa tested at doses of 10(8) CFU per eye induced corneal perforation and/or phthisis in C3H/HeN mice. The 50% infective doses of several strains were between 3 x 10(2) and 1 x 10(5) CFU per mouse eye. When this modified anesthetic procedure was used to evaluate the roles of different P. aeruginosa virulence factors in eye infections, pathology was not observed when eyes were inoculated with 10(8) CFU of strains deficient in production of a complete lipopolysaccharide or the RpoN sigma factor. A strain with a point mutation in the fur gene, involved in production of iron-regulated factors, showed decreased virulence, while a mutant deficient in both hemolytic and nonhemolytic phospholipase C was fully virulent. By modifying the anesthesia procedure, the corneal scratch model allows rapid evaluations of the roles of P. aeruginosa virulence factors in corneal infections.


Subject(s)
Corneal Diseases/microbiology , Pseudomonas aeruginosa/pathogenicity , Animals , Corneal Diseases/immunology , Female , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Virulence
12.
Infect Immun ; 62(8): 3485-93, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8039920

ABSTRACT

Pseudomonas aeruginosa is considered an extracellular pathogen. Using assays to determine intracellular survival in the presence of gentamicin, we have demonstrated that some strains of P. aeruginosa are able to invade corneal cells during experimental bacterial keratitis in mice. Although intracellular bacteria were detectable 15 min after inoculation, the number of intracellular bacteria increased in a time-dependent manner over a 24-h period. Levels of invasion were similar when bacteria were grown as a biofilm on solid medium and when they were grown in suspension. Intracellular bacteria survived in vitro for at least 24 h, although only minimal bacterial multiplication within cells was observed. P. aeruginosa PAK and Escherichia coli HB101 did not cause disease in this model and were not isolated from corneas after 24 h even when an inoculum of 10(8) CFU was applied. Transmission electron microscopy of corneal epithelium from eyes infected for 8 h revealed that intracellular bacteria were present within membrane-bound vacuoles, which suggests that bacterial entry was an endocytic process. At 24 h, the observation of many bacteria free in the cytoplasm indicated that P. aeruginosa was able to escape the endocytic vacuole. The ability of some P. aeruginosa strains to invade corneal epithelial cells may contribute to the pathogenesis or to the progression of disease, since intracellular bacteria can evade host immune effectors and antibiotics commonly used to treat infection.


Subject(s)
Cornea/microbiology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/pathogenicity , Animals , Bacterial Adhesion , Cornea/ultrastructure , Epithelium/microbiology , Keratitis/etiology , Mice , Mice, Inbred C57BL , Microscopy, Electron
13.
Infect Immun ; 62(5): 1799-804, 1994 May.
Article in English | MEDLINE | ID: mdl-8168942

ABSTRACT

To gain access to the corneal epithelium and cause infections keratitis, bacterial pathogens must first interact with ocular surface factors that could affect bacterial adherence. In this study, we demonstrated that the mucus layer, and, in particular, the mucin fraction of mucus, modulated adherence to intact corneal epithelium of Pseudomonas aeruginosa but not that of Staphylococcus aureus or Streptococcus pyogenes. Removal of endogenous mucus from rat or rabbit eyes increased the adherence of P. aeruginosa by 3- to 10-fold. Ocular mucus obtained from rat eyes, porcine stomach mucin, or bovine submaxillary gland mucin inhibited adherence of P. aeruginosa to uninjured corneal epithelium. The mucin fraction of ocular mucus, purified by ultracentrifugation, was found to contain the inhibitory activity, and inhibition was demonstrated at concentrations of mucin as low as 35 micrograms/ml. Ocular mucin was the only material tested that inhibited adherence of P. aeruginosa to an injured cornea. However, the binding of P. aeruginosa to immobilized substrates in vitro did not predict which fraction would possess antiadherence activity: bacteria bound well to whole ocular mucus, mucin, the nonmucin fraction of ocular mucus, and dilute human tears as well as to porcine stomach mucin and bovine submaxillary gland mucin. The effectiveness of the mucin fraction of ocular mucus at inhibiting the binding of P. aeruginosa to the cornea implies that this material is a barrier that protects the surface of the eye from P. aeruginosa adherence.


Subject(s)
Bacterial Adhesion , Cornea/microbiology , Mucus/physiology , Pseudomonas aeruginosa/pathogenicity , Animals , Female , Humans , Keratitis/etiology , Male , Rabbits , Rats , Rats, Wistar
15.
Invest Ophthalmol Vis Sci ; 31(4): 689-95, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2335437

ABSTRACT

It is generally believed that plasma membrane glycoconjugates influence corneal epithelial cell migration after wounding. Previous studies have focused on the role of glycoproteins in this event. The present study was designed to determine whether migration-specific glycolipids are synthesized by epithelium of healing rabbit corneas. Migrating and nonmigrating rabbit corneal epithelia were incubated with [3H]-galactose in an organ culture system for 48 hr. At the end of the labeling period, a neutral glycosphingolipid (NGSL) fraction was isolated from each radiolabeled epithelium and was analyzed by thin-layer chromatography. Three radiolabeled NGSL components, M1, M2 and M3 (M1-M3), were present in significantly higher amounts in the extracts of migrating as compared to nonmigrating epithelium. Chromatographic mobility of M3 was similar to that of a standard glucosylceramide; M1 and M2 migrated more slowly than M3. For characterization of the migration-related NGSL, a large amount of the starting material is required. Experiments, therefore, were conducted using cell cultures of rabbit corneal epithelium. Confluent (nonmigrating) cell cultures of rabbit corneal epithelium were found to synthesize either minimal or undetectable amounts of NGSL M1-M3. In contrast, we found that the NGSL M1-M3 are synthesized as major components by sparse (migrating) corneal epithelial cell cultures. Components M1-M3 were synthesized as major components by sparse cultures even in the absence of cell mitosis. This suggests that the increased synthesis of components M1-M3 by sparse cell cultures may be related to cell migration rather than cell mitosis.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cornea/metabolism , Glycolipids/metabolism , Animals , Cell Movement , Cells, Cultured , Chromatography, Thin Layer , Cornea/cytology , Cornea/physiology , Epithelial Cells , Epithelium/metabolism , Epithelium/physiology , Fluorouracil/pharmacology , Organ Culture Techniques , Rabbits , Wound Healing
16.
Infect Immun ; 58(1): 114-8, 1990 Jan.
Article in English | MEDLINE | ID: mdl-2104595

ABSTRACT

35S-labeled Pseudomonas aeruginosa isolates were shown to bind to neutral glycosphingolipids (NGSLs) of rabbit corneal epithelia in culture by a thin-layer chromatogram overlay procedure. The lipids of the corneal epithelial cells grown in culture were extracted and partitioned into a chloroform-rich lower phase containing NGSLs and an aqueous upper phase containing gangliosides. By using a dot-blot assay, at least six times more radiolabeled P. aeruginosa isolates were shown to bind to the lipids in the lower phase compared with those in the upper phase. Thin-layer chromatography of the lower-phase lipids followed by staining with an orcinol spray revealed at least 10 NGSL components and several fast-migrating, nonglycosylated neutral lipid components (including cholesterol). 35S-labeled P. aeruginosa was shown to bind to NGSL components 1, 2, 5, 6, and 9. P. aeruginosa-reactive NGSL components 6 and 9 migrated with chromatographic mobilities similar to those of the standards ceramide trihexoside (CT) and ceramide monohexoside, respectively. Components 1 and 2 migrated slightly ahead of asialo GM1, and component 5 migrated faster than globoside but slower than CT. Among the various standards tested, P. aeruginosa bound to asialo GM1 and, to a lesser extent, to ceramide dihexoside and CT but not to GM1, GD1A, GM3, or ceramide monohexoside. It remains to be determined whether any of the five P. aeruginosa-reactive NGSL components of corneal epithelium identified in this study plays a role in the development of corneal infection. However, we have previously shown that component 9, one of the five P. aeruginosa-reactive NGSL components identified in this study, is present in significantly greater amounts in migrating epithelia than it is in nonmigrating epithelia (N. Panjwani, G. Michalopoulos, J. Song, G. Yogeeswaran, and J. Baum, Invest. Ophthalmol. Vis. Sci., in press). This may prove to be of biological significance because it is generally believed that traumatized (migrating) epithelia are more susceptible to infection than normal (nonmigrating) epithelia are.


Subject(s)
Cornea/microbiology , Glycosphingolipids/metabolism , Pseudomonas aeruginosa/metabolism , Animals , Bacterial Adhesion , Cholesterol/metabolism , Chromatography, Thin Layer , Cornea/analysis , Epithelium/analysis , Epithelium/microbiology , Glycosphingolipids/analysis , In Vitro Techniques , Pseudomonas aeruginosa/pathogenicity , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...