J Neuropathol Exp Neurol Copyright Ó 2009 by the American Association of Neuropathologists, Inc. Vol. 68, No. 6 June 2009 pp. 677Y690 ORIGINAL ARTICLE OmpA Is the Critical Component for Escherichia coli InvasionYInduced Astrocyte Activation Hsueh-Hsia Wu, MS, Yi-Yuan Yang, PhD, Wen-Shyang Hsieh, MS, Chi-Hsin Lee, BS, Sy-Jye C. Leu, PhD, and Mei-Ru Chen, PhD From the Graduate Institute and Department of Microbiology, College of Medicine, National Taiwan University (H-HW, M-RC); School of Medical Laboratory Science and Biotechnology, College of Medicine, Taipei Medical University (H-HW, Y-YY); Department of Laboratory Medicine, Mackay Memorial Hospital (W-SH); and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University (C-HL, S-JCL), Taipei, Taiwan. Send correspondence and reprint requests to: Mei-Ru Chen, PhD, Graduate Institute and Department of Microbiology, College of Medicine, National Taiwan University, No 1, 1st section, Jen-Ai Road, Taipei 100, Taiwan; E-mail: [email protected] Supported by Grant Nos. NSC92-2320-B-038-056 (to H-HW) and NSC952320-B-002-087-MY3 (to M-RC) from National Science Council and Grant No. 9453 (to W-SH) from Mackay Memorial Hospital. Online-only color figures are available at http://www.jneuropath.com. ated with high mortality and morbidity. Most importantly, approximately half of the survivors display adverse neurologic complications. The pathologic complications of bacterial meningitis include cerebritis, brain abscess, empyema, and ventriculitis in the acute phase and the sequelae of cerebral atrophy and hydrocephalus (1). Neuronal injury associated with bacterial central nervous system (CNS) infection involves multiple microbial and host factors. For an understanding of the pathogenesis of these complications, the interactions between bacterial components and host cell responses in the CNS need to be elucidated. Escherichia coli strains with the K1 capsular polysaccharide are the most predominant Gram-negative bacteria associated with neonatal bacterial meningitis (2). The meningitis-associated E. coli K1 strain can translocate from the bloodstream to the CNS without disrupting the integrity of the blood-brain barrier (BBB) (3). A high level of bacteremia and invasion through brain microvascular endothelial cells (BMECs) seem to be determining factors that contribute to CNS infection (3). Several K1-associated components participate in BMEC binding and invasion; these include Fim H (4), K1 capsule (5), and outer membrane protein A (OmpA) (6Y8). Outer membrane protein A is one of the major outer membrane proteins of E. coli and plays important roles in maintaining the integrity of outer membrane and in bacterial conjugation (9Y11). It is also the receptor for several bacteriophages (12Y15). Outer membrane protein A is encoded by a 1038-bp open reading frame that consists of a 21-aminoacid leader peptide and the mature 325-amino-acid protein. The N-terminal membraneYanchoring domain forms an antiparallel A-barrel, which has 8 transmembrane A-strands connected by 3 short periplasmic turns and 4 relatively large, surface-exposed hydrophilic loops; the C-terminal domain interacts with the peptidoglycan layer in the periplasm to maintain outer membrane integrity (16). Outer membrane protein A is highly conserved through the evolution of Gram-negative bacteria and is important for the binding of E. coli to, and the invasion of, BMECs (17). Beyond the BBB, the most abundant cells in the CNS are astrocytes that provide physical and nutritional support for neurons. CNS injury results in astrocyte proliferation and morphologic changes with enhanced expression of glial fibrillary acidic protein (GFAP), the major constituent of glial intermediate filaments in mature astrocytes (18, 19). Astrocytes J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 677 Abstract Escherichia coli is the major Gram-negative bacterial pathogen in neonatal meningitis. Outer membrane protein A (OmpA) is a conserved major protein in the E. coli outer membrane and is involved in several host-cell interactions. To characterize the role of OmpA in the invasion of astrocytes by E. coli, we investigated OmpA-positive and OmpA-negative E. coli strains. Outer membrane protein A+ E44, E105, and E109 strains adhered to and invaded C6 glioma cells 10- to 15-fold more efficiently than OmpA-negative strains. Actin rearrangement, protein tyrosine kinase, and phosphoinositide 3Ykinase activation were required for OmpA-mediated invasion by E. coli. In vitro infection of C6 cells and intracerebral injection into mice of the E44 strain induced expression of the astrocyte differentiation marker glial fibrillary acidic protein and the inflammatory mediators cyclooxygenase 2 and nitric oxide synthase 2. After intracerebral infection with E44, all C57BL/6 mice died within 36 hours, whereas 80% of mice injected with E44 premixed with recombinant OmpA protein survived. Astrocyte activation and neutrophil infiltration were reduced in brain tissue sections in the mice given OmpA. Taken together, these data suggest that OmpAmediated invasion plays an important role in the early stage of E. coliYinduced brain damage, and that it may have therapeutic use in E. coli meningitis. Key Words: Astrocyte, Cyclooxygenase 2, Escherichia coli, Glial fibrillary acidic protein, Nitric oxide synthase 2, Outer membrane protein A. INTRODUCTION Despite advances in anti-microbial treatments and intensive care, bacterial meningitis remains a disease associ- Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. Wu et al J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 are also involved in intracerebral immune regulation (20). Activated astrocytes express cyclooxygenase 2 (COX-2) and nitric oxide synthase 2 (NOS2) (21, 22), which may subsequently lead to damage of neurons and glial cells, activation of BMECs, and overamplified inflammation responses (23Y26). The initial response of astrocytes to E. coli infection and the critical bacterial component(s) involved are not presently known. To provide insight into the pathogenic mechanisms of inflammation of the CNS, we determined whether OmpA protein mediates the invasion of astrocytes by E. coli. The invasion-induced activation of astrocytes was demonstrated by the expression of GFAP, COX-2, and NOS-2. The effects of recombinant OmpA protein on intracerebral E. coli challenge in C57BL/6 mice were also examined. with individual bacterial strains at multiplicity of infection (moi) of 0.1, 1, or 10 for 2 hours at 37-C, or at moi 10 for 0.5, 1, 1.5, 2, or 3 hours. Monolayers of cells were washed with culture medium 3 times and lysed in 0.5% Triton X100. The released bacteria were enumerated by plating on sheep blood agar plates (BAPs; Difco Laboratories). Assays were performed in duplicate and repeated at least 3 times. For intracellular bacteria assays, confluent monolayer cells (grown in 6-well plates) were incubated with bacteria at moi of 0.1, 1, or 10 for 2 hours at 37-C or at moi 10 for 0.5, 1, 1.5, 2, or 3 hours. The cells were washed with culture medium 3 times and incubated with culture medium containing 100 Kg/ml of gentamicin (Life Technologies, Gaithersburg, MD) for 2 hours to kill the extracellular bacteria. The monolayer cells were then washed 3 times and lysed in 0.5% Triton X-100. The released bacteria were enumerated by plating on BAP. To examine the signaling pathways involved in the invasion process, C6 cells were pretreated with individual inhibitor (Cytochalasin D, genistein, or LY294002, all from Calbiochem, San Diego, CA) at different concentrations for 1 hour before infection. Cytochalasin D was dissolved in sterilized water, whereas genistein and LY294002 were dissolved in dimethyl sulfoxide. Assays were performed in duplicate and repeated at least 3 times. MATERIALS AND METHODS Cell Line C6, a rat glioma cell line (ATCC CCL-107), was purchased from Bioresource Collection and Research Center, Hsinchu, Taiwan. Cells were cultured in Ham F10 medium with 15% horse serum, 2.5% fetal bovine serum, and 50 U/ml of penicillin-streptomycin (all from Life Technologies, Gaithersburg, MD). Chemicals, Bacteria, and Culture Medium Chemicals were all purchased from Sigma (St. Louis, MO) unless otherwise indicated. All Escherichia coli strains were kindly provided by Dr. K.S. Kim (Division of Pediatric Infectious Diseases, School of Medicine, Johns Hopkins University, Baltimore, MD); they are summarized in Table 1. E44 is K1 strain RS218 (O18:K1:H7) isolated from the cerebrospinal fluid of a neonate with meningitis (17). E91 is an E44 mutant lacking the entire ompA gene. E105 is an OmpA complemented mutant of E91 that carries pRD87 (which contains the ompA gene on pUC9). E111 is E91 with pUC9 vector control. E109 was obtained by transformation of E91 with a pRD87 derivative that lacks the C-terminal 53 amino acids of OmpA. MG1655 is a nonpathogenic strain that is noninvasive for the BBB. Bacteria were grown in brain heart infusion broth with appropriate antibiotics (all from Difco Laboratories, Detroit, MI). For infection experiments, overnight cultures were expanded in brain heart infusion broth and incubation at 37-C for 2 to 3 hours to mid-log phase. Bacteria were centrifuged and resuspended in cell culture medium without antibiotics. Mouse Strain C57BL/6 mice were obtained from the National Laboratory Animal Center of Taiwan and kept under pathogen-free conditions. Animal procedures were performed in accordance with an approved protocol (LAC-96-0031) under the institutional protocol of Taipei Medical University. Adhesion and Invasion Assays For cell-associated bacteria studies, confluent cultures of C6 cells grown in 6-well plates (Corning, NY) were incubated 678 Intracellular Bacteria Survival Confluent monolayer cells grown in 6-well plates were infected with bacteria at moi 10 for 2 hours. Cells were washed 3 times and then incubated with culture medium containing 100 Kg/ml of gentamicin for 2 hours to kill the extracellular bacteria. The cells were then incubated with medium containing 20 Kg/ml of gentamicin for 2, 4, 24, or 48 hours, and recoverable colony-forming units (CFUs) were determined on BAPs. Assays were performed in duplicate and repeated at least 3 times. Electron Microscopy Confluent cultures of C6 cells grown in 2-well culture slides were incubated with bacteria at moi 10 for 30 or TABLE 1. Adhesion and Invasion of E. coli Strains into C6 Glioma Cells Mean CFU (TSD)/Well E. coli Strain E44 E91 E105 E109§ E111 MG1655 Relevant Characteristic(s)* K1 RS218 spontaneous mutant E44 OmpAj E91 OmpA+ E91 truncated OmpA+ E91 OmpAj K12 laboratory strain Adhesion Invasion (6.4 T 0.1) 107 (4.5 T 0.3) 104 (5.2 T 0.4) 106† (6.7 T 0.3) 107 (6.7 T 0.2) 107 (5.4 T 0.5) 106† (9.0 T 1.5) 106† (6.2 T 0.4) 103‡ (4.6 T 0.3) 104 (4.6 T 0.1) 104 (6.1 T 0.2) 103† (5.7 T 0.5) 103† SD, standard deviation. *Specific characteristics of these strains are described in Reference 17. †p G 0.01 compared with E44 by Student t test. ‡p G 0.05 compared with E44 by Student t test. §C-Terminal 53 amino acids of OmpA are deleted in this strain. Ó 2009 American Association of Neuropathologists, Inc. Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 OmpA in E. coli Invasion Into Astrocytes 90 minutes at 37-C, washed 3 times with prewarmed phosphate-buffered saline (PBS), and fixed with 2.5% glutaraldehyde. For transmission electron microscopy, samples were fixed with 2% osmium tetroxide for 2 hours, washed 3 times with PBS, dehydrated in a gradient series of alcohol concentrations, and embedded in Epon 812 (Serva, Heidelberg, Germany). Ultrathin sections were stained with uranyl acetate and lead citrate and examined with an EM 906 transmission electron microscope (Zeiss). were purified with Ni2+-charged sepharose according to the manufacturer’s instructions (Amersham). His-OmpA and His-enolase were eluted by an elution buffer (20 mmol/L of sodium phosphate, 500 mmol/L of NaCl, and 500 mmol/L of imidazole). The samples were massively dialyzed against the elution buffer without imidazole then a PBS (pH 7.4) to reduce the salt concentrations. Electrophoresis and Immunoblotting Electrophoresis was performed on 10% sodium dodecyl sulfateYpolyacrylamide gel electrophoresis (SDS-PAGE). Reagents for SDS-PAGE were from Bio-Rad (Hercules, CA). After electrophoresis, proteins on the gel were electrotransferred onto a polyvinylidene fluoride (PVDF) membrane (Amersham Pharmacia Biotech, Buckinghamshire, UK). After transfer, the PVDF membranes were blocked with blocking solution containing 5% skim milk in TBST (10 mmol/L Tris, pH7.5, 100 mmol/L NaCl and 0.1% Tween 20) for 1 hour at room temperature. Antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA) unless otherwise indicated. The PVDF membrane was incubated with a solution containing mouse anti-GFAP antibody (1:100 diluted), rabbit antiYCOX-2 antibody (1:200), rabbit antiYNOS-2 antibody (1:200), or goat anti-actin antibody (1:200) in the blocking buffer for 2 hours. After washing, the PVDF membrane was incubated with peroxidase-linked secondary antibody (1:2000) for 1 hour. Finally, the PVDF membrane was developed using a chemiluminescence kit (Amersham). Immunofluorescence Staining and Confocal Analysis Cryosections of mouse brains or slide-cultured C6 cells were fixed with 4% paraformaldehyde. Samples were blocked with 1% bovine serum albumin and then incubated with a solution containing mouse anti-GFAP antibody (1:50), rabbit antiYCOX-2 antibody (1:50), rabbit antiYNOS-2 antibody (1:50), rat antiYLy-6G antibody (1:100; eBioscience, San Diego, CA), or goat antiYE. coli antibody (1:100; Abcam, Cambridge, UK) for 1 hour. After washing, samples were incubated with fluorescein isothiocyanate (FITC)Y labeled secondary antibody (1:200) or rhodamine-labeled secondary antibody (1:50) for 1 hour. After washing, propidium iodide or Hoechst stain was used to stain DNA. Slides were mounted in 50% glycerol-PBS and then examined with the TCS SP5 Confocal Spectral Microscope Imaging System (Leica). Expression and Purification of His-OmpA and His-Enolase Protein The DNA fragment containing full-length ompA and enolase gene was amplified with polymerase chain reaction using restriction enzyme sites containing primers, digested with SacI and XhoI, and ligated into pET-21 expression vector (Novagen, Darmstadt, Germany). The resultant plasmid was transformed into E. coli BL-21 (DE3) strain. Protein expression was induced by 0.5 mmol/L of isopropyl-A-Dthiogalactopyranoside. His-OmpA and His-enolase proteins Ó 2009 American Association of Neuropathologists, Inc. Animal Experiments C57BL/6 mice 8 to 12 weeks old were randomly distributed into groups. To assess the survival of C57BL/6 mice after intracerebral bacterial administration, the protocol developed by Tsao et al (23) was followed. Each group of 5 mice was anesthetized with pentobarbital sodium salt (50 mg/kg) by intraperitoneal injection, and then each brain was infected with bacteria 5 105 in 20 Kl of PBS, or 5 Kg of lipopolysaccharide (LPS; from E. coli O26:B6, Sigma) in 20 Kl of PBS by intracerebral injection. Phosphate-buffered saline (20 Kl) was used as a negative control. The mice were monitored for survival every 12 hours for 8 days. To investigate the role of recombinant OmpA protein in the survival of C57BL/6 mice after intracerebral E44 administration, each brain was infected with E44 5 105 in 20 Kl of PBS by intracerebral injection in the absence or presence of OmpA (8 Kg or 20 Kg) or 20 Kg enolase. Phosphate-buffered saline (30 Kl) was used as a negative control. Survival of C57BL/6 mice was observed up to 8 days postadministration. For immunofluorescence staining and Western blotting, groups of 3 C57BL/6 mice were anesthetized and then infected with bacteria by intracerebral injection. Mice were killed and brains were removed at 24 hours postchallenge. For immunofluorescence staining, brains were embedded in OCT compound, and 6-Km cryosections were cut. For Western blotting, brains were homogenized with protein extraction buffer (50 mmol/L of Tris HCl, pH 8.0, 150 mmol/L of NaCl, 1% NP-40, and the protease inhibitor cocktails [Bio-Rad], including 0.2 mmol/L of PMSF, 20 Kg/ml of aprotinin, and 20 Kg/ml of leupeptin) and harvested for whole cell lysate. To detect remnant bacteria in the brain, groups of 3 C57BL/6 mice were infected with bacteria by intracerebral injection. Mice were killed at various times postchallenge. The brains were aseptically removed and homogenized with 3% gelatin in PBS. The samples were serially diluted, and CFU was determined on BAP. Statistics The results were expressed as the viation of 3 independent experiments. differences between treatment groups Student t test. Results were considered calculated p value was less than 0.05. mean T standard deThe significance of was determined by significant when the RESULTS OmpA of E. coli K1 Contributes to Adhesion to and Invasion of C6 Cells To provide an in vitro system for E. coli invasion of astrocytes, we chose rat glioma C6 cells (27, 28). Pilot 679 Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. Wu et al J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 experiments were performed with E44 (OmpA+), E91 (OmpA-), and a laboratory nonpathogenic control strain, MG1655, to determine the optimal conditions for adhesion and invasion. Monolayer cultured C6 cells were incubated with bacteria at moi of 0.1, 1, or 10 for 2 hours. The cellassociated E. coli increased in a dose-dependent manner (Fig. 1A). The effect of incubation time was then monitored at moi 10. The results showed that attachment of the E. coli strains increased steadily up to 3 hours (Fig. 1B). Next, the invasion abilities of E44, E91, and MG1655 were monitored by incubating bacteria at moi of 0.1, 1, or 10 with C6 cells for 2 hours (Fig. 1C) or at moi 10 for up to 3 hours (Fig. 1D). Both the adhesion and invasion abilities of E44 are much greater than those of E91 and MG1655. Because all strains displayed dose-dependent and time-dependent invasion, moi 10- and 2-hour incubation were used subsequently as the FIGURE 1. Adhesion and invasion of C6 cells by E. coli strains. (A) For dose-dependent adhesion assays, confluent cultures of C6 cells were infected with bacteria at the indicated moi for 2 hours. Recoverable CFUs were determined on sheep BAPs. (B) For time-course adhesion assays, confluent cultures of C6 cells were infected with bacteria at the indicated moi 10 for the indicated time periods, and recoverable CFU were determined. (C) For dose-dependent invasion assays, confluent cultures of C6 cells were infected with bacteria at the indicated moi for 2 hours. Cells were incubated with medium containing 100 Kg/ml of gentamicin for 2 hours to kill extracellular bacteria. Recoverable CFUs were then determined. (D) For time-course invasion assays, confluent C6 cell cultures were infected with bacteria at moi 10 for the indicated time periods. Cells were then incubated with medium containing 100 Kg/ml of gentamicin for 2 hours to kill the extracellular bacteria and recoverable CFU determined. (E) To detect the survival of internalized bacteria, confluent C6 cell cultures were infected with bacteria at moi 10 for 2 hours. After washing, the culture was incubated with medium containing 100 Kg/ml of gentamicin for 2 hours to kill the extracellular bacteria and further incubated with medium containing 20 Kg/ml of gentamicin for the indicated periods. Recoverable CFUs were then determined. Data represent the average of 3 independent experiments; error bars indicate standard deviations. 680 Ó 2009 American Association of Neuropathologists, Inc. Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 OmpA in E. coli Invasion Into Astrocytes standard assay conditions for this study. We then determined whether OmpA influences the intracellular survival of E. coli. The results showed that internalized bacteria decreased dramatically at 24 hours postincubation and became undetectable at 48 hours postinfection (Fig. 1E). At the same time, the growth of C6 cells was not affected by the infection with E. coli, as indicated by viable cell number counts performed daily for up to 7 days postinfection (data not shown). To determine whether OmpA contributes to E. coli invasion into C6 cells, a panel of E. coli strains generated in E44 was used for adhesion and invasion assays. Among them, E91 is OmpA-deficient; E105 and E109 are E91 complemented by a plasmid that carries a full-length or truncated ompA, respectively; E111 contains the vector control (Table 1) (17). We observed that OmpA+ strains FIGURE 3. Signaling pathways involved in the invasion of E44 into C6 cells. Confluent cultures of C6 cells were pretreated for 1 hour before E44 infection with inhibitors and then infected with E44 (moi 10) for 2 hours and assayed for invasion. The inhibitors cytochalasin D (A), genistein (B), or LY294002 (C) were added at the indicated concentrations. Cytochalasin D was dissolved in sterilized water; genistein and LY294002 were dissolved in dimethyl sulfoxide. Results are shown as relative invasiveness with CFUs of untreated controls as 100%. Data represent the average of 3 independent experiments; error bars indicate standard deviations. **, p G 0.001; j, untreated control; 0, solvent control. FIGURE 2. Invasion of E44 into C6 cells under TEM. Confluent monolayers of C6 cells were infected with bacteria at moi 10 for 30 (A) or 90 minutes (B); ultrathin sections were then prepared for TEM (magnification: 8,000). TEM, transmission electron microscopy. Ó 2009 American Association of Neuropathologists, Inc. E44, E105, and E109 adhered to and invaded into C6 cells in numbers approximately 10- to 15-fold higher than the OmpA- strains, E91, and E111. E91 behaved similarly to MG1655 in adhesion and invasion assays (Table 1). Because the growth rates of various E. coli strains alone in culture medium were similar (data not shown), the data suggest that depletion of OmpA diminishes the adhesion 681 Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. Wu et al J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 and invasion abilities of E. coli, rather than affecting its growth per se. The ability of bacteria to invade into C6 cells was examined further by electron microscopy at 30 and 90 minutes post-E44 infection (Fig. 2A, B). The internalized bacteria were surrounded by a membrane-like structure and located within vacuoles. Intracellular E91 showed a similar pattern, but with far less frequency (data not shown). Invasion of E. coli was associated with the formation of lamellipodia of C6 cells (Fig. 2B), suggesting that FIGURE 4. Outer membrane protein AYmediated E. coli invasion induces GFAP expression in C6 glioma cells. Confluent cultures of C6 cells were infected with E coli (moi, 10) for 2 hours, followed by culture medium containing 100 Kg/ml of gentamicin for 24 hours. Lipopolysaccharide (1 Kg/ml) treatment for 24 hours served as a positive control. (A, B) Western analysis of whole cell lysate to detect GFAP and actin. (C) Immunofluorescence staining after infection of C6 cells grown on 22-mm glass coverslips using anti-GFAP antibody. DNA was stained with PI and examined by confocal microscopy. E44-infected and LPS-treated cells show enhanced GFAP immunoreactivity. (D) C6 cells were pretreated with 0.3 Kg/ml of CD, 100 Kmol/L of LY294002, or 100 Kmol/L of genistein for 1 hour before infection with E44. After 2 hours, the cells were washed and incubated with culture medium containing 100 Kg/ml of gentamicin for 24 hours. Whole-cell lysate was collected, and Western analysis was performed to detect GFAP and actin. C6 cells treated with inhibitor alone serve as a negative control. CD, cytochalasin D; PI, propidium iodine. 682 Ó 2009 American Association of Neuropathologists, Inc. Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 OmpA in E. coli Invasion Into Astrocytes macropinocytosis seems to be involved in the internalization process. Actin Rearrangement, Protein Tyrosine Kinase, and Phosphoinositide-3-Kinase Activation Are Required for OmpA-Mediated E. coli Invasion We next explored the cellular signaling pathways involved in the E44 invasion process. It has been reported FIGURE 6. Survival of C57BL/6 mice after infection with E. coli strains. Groups of 5 C57BL/6 mice were anesthetized and challenged with 5 105 bacteria or 5 Kg LPS by intracerebral injection. Phosphate-buffered saline was used as a negative control. The mice were monitored for survival up to 8 days. in the case of E. coli K1, Listeria monocytogenes, group B Streptococcus, and Moraxella catarrhalis that actin cytoskeleton rearrangement is a prerequisite of bacterial invasion (29Y33). To determine whether actin rearrangement is involved in E44 invasion, C6 cells were pretreated with the microfilament-depolymerizing agent cytochalasin D at concentrations of 0.01, 0.03, 0.1, or 0.3 Kg/ml for 1 hour before infection. Data obtained from invasion assays revealed that E44 invasion was blocked by cytochalasin D in a dosedependent manner, and greater than 95% inhibition was observed at 0.1 Kg/ml (Fig. 3A), suggesting that actin polymerization is required for OmpA-mediated E. coli invasion into C6 cells. Furthermore, invasion of E. coli K1 into BMECs depends on tyrosine phosphorylation of focal adhesion kinase (34). To determine whether protein tyrosine kinase activation is required for E44 invasion, C6 cells were pretreated with the general protein tyrosine kinase inhibitor genistein at increasing concentrations for 1 hour before infection. The results indicated that genistein blocked E44 FIGURE 5. Infection of OmpA+ E. coli induces astrocyte activation and the expression of COX-2 and NOS-2 in vivo. Groups of 3 C57BL/6 mice each were anesthetized and challenged with 5 105 bacteria or 5 Kg LPS by intracerebral injection. Phosphate-buffered saline was used as a negative control; the mice were killed at 24 hours postinfection. (A, B) Confocal images of immunofluorescence staining using mouse anti-GFAP, rabbit antiYCOX-2, or rabbit antiYNOS-2 antibody, followed by FITC-conjugated anti-mouse IgG or rhodamineconjugated anti-rabbit IgG antibody. DNA was stained with Hoechst. (C, D) Western analysis of whole cell lysates detects COX-2 or NOS-2 and actin. (E) Confocal images of consecutive brain cryosections of E44-infected mouse brains at 24 hours postinfection stained with mouse anti-GFAP or goat antiYE. coli antibody, followed by FITC-conjugated anti-mouse IgG antibody or rhodamine-conjugated anti-goat IgG antibody. DNA was stained with Hoechst. IgG, immunoglobulin G. Ó 2009 American Association of Neuropathologists, Inc. 683 Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. Wu et al J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 FIGURE 8. His-OmpA protects C57BL/6 mice from intracerebral infection of E44. Groups of 5 C57BL/6 mice were infected with 5 105 E44 bacteria by intracerebral injection in the absence or presence (8 or 20 Kg) of His-OmpA or 20 Kg Hisenolase. Phosphate-buffered saline served as a negative control. Mice were monitored for survival at 12-hour intervals for 8 days. FIGURE 7. Effects of His-OmpA on E. coli invasion and GFAP expression in C6 glioma cells. (A) Purified recombinant OmpA and enolase proteins were displayed by 10% SDS-PAGE and stained with Coomassie blue. The expected molecular weight is 38 kDa for His-OmpA and 50 kDa for His-enolase. (B) Immunoblotting of recombinant OmpA protein. Chicken antiOmpA IgY (1:5000) and HRP-conjugated donkey anti-chicken IgY antibody (1:10000) were used. Immunoreactive bands were visualized with DAB. (C) Confluent C6 cell monolayers were infected with E44 (moi, 10) in the absence or presence (4 or 40 Kg/ml) of His-OmpA or 40 Kg/ml His-enolase for 2 hours. The cells were then incubated with medium containing 100 Kg/ml of gentamicin for 2 hours to kill the extracellular bacteria. After washing, recoverable CFUs were determined. Results are expressed as relative invasiveness with CFU of E44 administration as 100%. Data represent the average of 3 independent experiments; error bars indicate standard deviations. **, p G 0.001. Confluent C6 cell cultures were infected with E44 (moi, 10) in the presence (4 or 40 Kg/ml) or absence of His-OmpA for 2 hours. Western analysis of whole cell lysates was performed to detect GFAP and actin. (D) Confluent cultures of C6 cells were infected with E44 (moi, 10) in the absence or presence (40 Kg/ml) of His-OmpA or 40 Kg/ml Hisenolase for 2 hours. After washing, GFAP expression in whole cell lysates was detected by immunoblotting. >OmpA, antiOmpA antibody; DAB, diaminobenzidine; HRP, horseradish peroxidase; IgY, immunoglobulin Y. 684 invasion in a dose-dependent manner, and greater than 90% inhibition was observed at a concentration of 30 Kmol/L (Fig. 3B); this suggests that protein tyrosine kinase activation is also required for OmpA-mediated E. coli invasion. As shown in Figure 2A and B, bacteria seemed to be internalized through macropinocytosis that comprises a phosphoinositide-3 (PI 3)YkinaseYdependent contractile mechanism (35). To investigate the involvement of PI 3Ykinase during E44 invasion, C6 cells were pretreated with the PI 3Ykinase inhibitor LY294002 at 3, 10, 30, or 100 Kmol/L for 1 hour before infection. E44 invasion was blocked by LY294002 in a dose-dependent manner; greater than 90% inhibition was achieved at a concentration of 30 Kmol/L (Fig. 3C), indicating that PI 3Ykinase is also involved in OmpA-mediated E. coli invasion. Taken together, these results demonstrate that rearrangement of the actin cytoskeleton and activation of protein tyrosine kinase and PI 3Ykinase are all needed for OmpA-mediated E. coli invasion of C6 cells. OmpA-Mediated E. coli Invasion Activates C6 Glioma Cells To determine whether C6 cells are activated by E. coli infection, immunoblotting was used to detect GFAP levels. The infection of OmpA+ strains E44, E105, and E109 but not OmpA- strains E91, E111, and MG1655 induced GFAP expression (Fig. 4A and B). By confocal microscopy, infection of E44, but not E91 or MG1655, induced cytoplasmic accumulation of GFAP (Fig. 4C). These results suggest that E. coliYinduced GFAP expression is dependent on OmpA. Next, invasion inhibitors were tested for their effects on E. coliYinduced GFAP accumulation. C6 cells were pretreated with 0.3 Kg/ml of cytochalasin D, 100 Kmol/L of LY294002, Ó 2009 American Association of Neuropathologists, Inc. Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 OmpA in E. coli Invasion Into Astrocytes or 100 Kmol/L of genistein for 1 hour before infection with E44 and compared with mock-infected cells. Glial fibrillary acidic protein expression induced by E44 was blocked sig- nificantly in the presence of cytochalasin D, LY294002, or genistein (Fig. 4D); these results suggest that OmpA-mediated invasion is necessary for activation of C6 cells. FIGURE 9. His-OmpA inhibits the activation of astrocytes and the expression of COX-2 and NOS-2 in C57BL/6 mice brains after E44 infection. Groups of 3 C57BL/6 mice were anesthetized and infected with 5 105 of E44 by intracerebral injection in the presence (8 or 20 Kg) or absence of His-OmpA. Phosphate-buffered saline served as a negative control. Mice were killed, and brains were removed at 24 hours postinfection. (A, B) Immunofluorescence staining with mouse anti-GFAP, rabbit antiYCOX-2, or rabbit antiYNOS-2 antibody, followed by FITC-conjugated anti-mouse IgG antibody or rhodamine-conjugated anti-rabbit IgG antibody. Hoechst was used for DNA staining. (C, D) Brain whole cell lysates were analyzed for COX-2 or NOS-2 and actin by immunoblotting. Ó 2009 American Association of Neuropathologists, Inc. 685 Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. Wu et al J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 Intracerebral Injection of OmpA+ E. coli Induces the Activation of Astrocytes and the Expression of COX-2 and NOS-2 periments, groups of mice were infected by intracerebral injection with 5 103, 5 105, or 5 107 bacteria. The 100% lethal dose of E44 was 5 105, and the 100% lethal dose of E91 and MG1655 was 5 107 (data not shown). The infectious inoculums used were 5 105 in this study. Groups of 3 C57BL/6 mice were anesthetized and then infected with of E44, E91, MG1655, or 5 Kg of LPS by intracerebral injection. At 24 hours postchallenge, the mice were killed, and brains were removed and embedded in OCT for cryosectioning. Because inflammatory molecules COX-2 We then determined whether OmpA is required for activation of astrocytes by E. coli in vivo. Because the invasive capability of E44 was 25- to 50-fold greater than that of E91 in BMECs (17), we challenged mice by intracerebral injection to avoid interference of the BBB according to the protocol of Tsao et al (23). In pilot ex- FIGURE 10. His-OmpA reduces neutrophil infiltration in C57BL/6 mouse brain after E44 infection. Consecutive brain cryosections used in Figures 5 and 9 from mice that had been challenged with PBS (control), E44 (5 105), E44 and 20 Kg His-OmpA, E91, MG1655, or 5 Kg LPS were stained with rat antiYLy-6G antibody and FITC-conjugated secondary antibody to demonstrate neutrophil infiltration. Hoechst was used for DNA staining; the slides were observed with confocal microscopy. Bar = 25 Km. 686 Ó 2009 American Association of Neuropathologists, Inc. Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 and NOS-2 mediate a large amount of the neurologic damage in CNS infections (23, 26), immunofluorescence staining was performed and observed by confocal microscopy to detect GFAP, COX-2, and NOS-2 expression in the brains after infection. As shown in Figure 5A, E44 infection induced accumulation of GFAP and expression of COX-2. The overlay of both signals indicated that COX-2 expression was colocalized with GFAP in activated astrocytes. In parallel, infection of E44 also induced NOS-2 expression in astrocytes expressing high levels of GFAP (Fig. 5B). Neither the activation of astrocytes nor the expression of COX-2 or NOS-2 was observed in E91- or MG1655-treated animals (Fig. 5A, B). Furthermore, groups of 3 mice each were infected with bacteria or 5 Kg of LPS by intracerebral injection and analyzed for COX-2 and NOS-2 expression by immunoblotting. Cyclooxygenase 2 and NOS-2 expression were induced after infection with E44, but not E91 or MG1655 (Fig. 5C, D). Furthermore, the consecutive brain cryosections of E44-infected mouse brains at 24 hours postinfection were costained with mouse anti-GFAP antibody and goat antiYE. coli antibody. Colocalization of bacteria and GFAP was observed in some areas with residual bacteria (Fig. 5E). Taken together, these results suggest that OmpAmediated E. coli infection activates astrocytes and induces the expression of COX-2 and NOS-2 in the mouse brain. Intracerebral Injection of OmpA+ E. coli Strain Causes the Death of C57BL/6 Mice To identify the role of OmpA in the development of E. coliYinduced CNS infection in vivo, groups of 5 mice were infected with E. coli K1 strains (E44, E91, E105, E109, and E111), MG1655, or 5 Kg of LPS by intracerebral injection and assessed for 8 days. Mice died within 48 hours of intracerebral injection of the OmpA+ strains E44, E105, or E109, whereas mice challenged with the OmpA- strains E91 or E111, MG1655, or with LPS lived for 8 days before they were killed (Fig. 6). This suggests that OmpA-mediated E. coli infection subsequently leads to the death of the mice. Recombinant OmpA Protein Inhibits E. coli Invasion and Activation of C6 Cells Because purified OmpA protein inhibited the invasion of BMECs by OmpA+ E. coli (8, 17), bacterially expressed recombinant His-OmpA and the negative control His-enolase were purified. Escherichia coli enolase is a glycolytic enzyme that catalyzes 2-phosphoglycerate into phosphoenolpyruvate (36). The homogeneities of the purified His-OmpA and His-enolase were revealed by Coomassie blue staining and confirmed by immunoblotting (Fig. 7A, B). The purified His-OmpA and His-enolase displayed molecular weights of 38 and 50 kDa on 10% SDS-PAGE, as predicted. To examine the effect of His-OmpA on E44 invasion, C6 cell monolayers were incubated with E44 in the absence or presence of His-OmpA. There was approximately 50% inhibition of invasion with 4 Kg/ml of His-OmpA and approximately 83% inhibition with 40 Kg/ml of His-OmpA; no inhibitory effect was observed in the presence of Hisenolase (Fig. 7C). By immunoblotting, the E44-induced GFAP expression was suppressed significantly by His-OmpA Ó 2009 American Association of Neuropathologists, Inc. OmpA in E. coli Invasion Into Astrocytes TABLE 2. Viable Bacteria in Brain Tissue After Intracerebral Injection of E. coli Mean CFU (TSD)/Brain in E. coli Strains Tested Time After Infection, hours 12 24 48 E44 + Recombinant OmpA E44 6 (2.3 T 0.4) 10 (2.4 T 0.1) 107 (6.0 T 0.3) 107 5 (2.0 T 0.3) 10 ND ND E91 (1.8 T 0.3) 105 ND ND Groups of mice (n = 3 per group) were given intracerebral injections of 5 105 E. coli. The brains were collected at the indicated time points after injection, and the bacteria remaining in the brains were quantified. ND, not detectable. in a dose-dependent manner (Fig. 7C). In contrast, Hisenolase did not inhibit E44-induced GFAP expression (Fig. 7D). These data indicate that recombinant OmpA can specifically prevent bacterial invasion and the subsequent accumulation of GFAP in C6 cells. Recombinant His-OmpA Protects C57BL/6 Mice from E44-Induced Death We next determined whether the presence of HisOmpA also protects mice from the death caused by intracerebral injection of E44. Groups of 5 C57BL/6 mice were challenged by intracerebral injection with E44 alone or a mixture of E44 and 8 or 20 Kg of His-OmpA or 20 Kg of His-enolase, and survival of the mice was assessed for 8 days. The mice challenged with E44 alone or E44 + 20 Kg of His-enolase died within 36 hours, whereas 40% of mice survived after the injection of E44 + 8 Kg of His-OmpA, and 80% of mice with E44 + 20 Kg of His-OmpA to 8 days postinfection (Fig. 8). Thus, recombinant His-OmpA can protect C57BL/6 mice from intracerebral infection by E44. Recombinant His-OmpA Inhibits the InfectionInduced Activation of Astrocytes and the Expression of COX-2 and NOS-2 In Vivo To determine whether recombinant His-OmpA affects E44-induced activation of astrocytes and the expression of COX-2 and NOS-2 in vivo, groups of 3 C57BL/6 mice were infected with E44 alone or a mixture of E44 and 20 Kg of His-OmpA. Mice were killed at 24 hours postchallenge, and the brains were removed, embedded in OCT for cryosectioning, and the sections were examined by confocal microscopy. The presence of His-OmpA reduced the E44-induced expression of GFAP, COX-2, and NOS-2 in the mouse brains (Fig. 9A, B). Further groups of mice were infected with E44 alone or a mixture of E44 and 8 or 20 Kg of HisOmpA. At 24 hours postchallenge, the brains were removed, homogenized with protein extraction buffer, and analyzed in immunoblotting. The results confirmed the expression of COX-2 and NOS-2 was inhibited specifically by His-OmpA in a dose-dependent manner (Fig. 9C, D). Consecutive brain sections were stained with antibody Ly-6G to observe neutrophil infiltrates in the forebrain region. Consistent with the survival experiment in Figure 6, intracerebral injection of E44, but not E91, MG1655, or LPS, induced neutrophil infiltration near the major blood vessels. In contrast, the 687 Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. Wu et al J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 mixture of 20 Kg of His-OmpA with E44 reduced the numbers of neutrophils in the forebrain region (Fig. 10). Simultaneously, the remnant bacteria in brain infected with E44 increased up to (2.4 T 0.1) 107 at 24 hours postinfection and (6.0 T 0.3) 107 at 48 hours postinfection; bacteria in brains infected with E91 or E44 with recombinant OmpA were undetectable at 24 hours postinfection (Table 2). These results demonstrate that mouse survival correlates with bacterial clearance. Taken together, these data indicate that His-OmpA can protect astrocytes from E44-induced activation, the expression of COX-2 and NOS-2, and neutrophil infiltration in mouse brains, effects that may protect the mice from severe neuronal damage and death. OmpA experiments also demonstrate that OmpA protein, but not the amount of Fim protein, contributes to the invasion of E. coli into astrocytes. Several host cell pathways are involved in the invasion of E. coli K1 into BMECs, although they are not fully elucidated. For example, rearrangement of the actin cytoskeleton was shown to be a prerequisite for BMEC invasion (30), and tyrosine phosphorylation of focal adhesion kinase and PI 3Ykinase activation are also required for E. coli invasion into BMECs (34, 44). The intracellular signaling pathways required for other microorganisms to invade into host cells are diverse. For example, rearrangement of the actin cytoskeleton was also shown to be a prerequisite for BMEC invasion by L. monocytogenes and group B Streptococcus (31, 32) and for invasion of pulmonary epithelial cells by M. catarrhalis (33). Invasion of BMEC by L. monocytogenes and of epithelial cells by M. catarrhalis, however, does not require phosphorylation of focal adhesion kinase (33, 34). Activation of PI 3Ykinase also is involved in L. monocytogenes invasion of BMEC and epithelial cells and M. catarrhalis invasion into pulmonary epithelial cells (33, 34, 45), in contrast, invasion of BMEC by group B Streptococcus was independent of PI 3Ykinase activation (3). Our present data indicate that rearrangement of the actin cytoskeleton, protein tyrosine kinase, and PI 3Ykinase activation are all required for E44 invasion of C6 cells. Thus, the invasion of bacteria into C6 cells may reflect the outcome of macropinocytosis, which is usually induced by the interaction of microbial factors with nonphagocytic cells, and followed by the activation of tyrosine kinases and actin-mediated ruffling. This idea is supported further by our electron micrographs showing that adherence of E. coli to C6 cell was accompanied by the formation of membrane ruffles and lamellipodia (Fig. 2B). Escherichia coli was then internalized and located within intracellular vacuoles, suggesting that a trigger-like uptake mechanism is involved in this process. Nevertheless, some of the macropinosome markers such as early endosomal autoantigen 1 or transferrin receptor for early endosome and Rab7 or Lamp 1 for late endosome and late endosome/lysosome (46) should be investigated to clarify the mechanisms involved in the E44 invasion of C6 cells in the future. We observed that internalized bacteria decreased dramatically at 24 hours postincubation in gentamicincontaining medium and became undetectable at 48 hours postinfection (Fig. 1E). To examine whether exocytosis or intracellular killing is mediated for bacteria clearance, we performed an experiment that C6 cells were incubated in antibiotic-free medium after E44 infection. At 24 hours postincubation, (6.5 T 2.1) 105 CFU/ml were recovered from the medium, suggesting that at least a portion of bacteria may be exocytosed after invasion (data not shown); it was not clear, however, whether intracellular killing occurred during this process. In the studies of BMECs, vacuoles containing E. coli K1 acquired markers for early endosome, late endosome, and late endosome/lysosome, but not the lysomal enzyme cathepsin D, suggesting E. coli K1 is able to direct the E. coli containing vacuoles to escape from fusion with lysosome and avoid the killing by lysosomal enzymes (7). Further study is required to understand the fate of internalized E44 in C6 cells. DISCUSSION We identified OmpA as the primary bacterial component involved in adhesion, invasion, and activation of astrocytes by E. coli. To our knowledge, this is the first report demonstrating that recombinant OmpA protein can protect astrocytes from activation induced by invasion of E. coli and, most importantly, protects the mice from death caused by intracerebral infection of OmpA+ E. coli. Outer membrane protein A was found to be the major component of E. coli that mediates host-cell interactions. In the noninvasive strain enterohemorrhagic E. coli, OmpA functions as an adhesion molecule that binds to HeLa, colonic epithelial and dendritic cells (37, 38). In invasive strains, OmpA is important for the invasion of E. coli K1 into macrophages and monocytes (39) and for binding and invasion into BMECs (7, 17, 40). We found that both the adhesion and invasion capacities of OmpA+ E. coli K1 strains (E44, E105, E109) were approximately 10to 15-fold greater than those of OmpAj E. coli K1 strains (E91, E111), that OmpA is the major determinant enabling E. coli to adhere to and invade into astrocytes, and that its binding is the most critical step for E. coli K1 invasion of C6 cells. Outer membrane protein A protein consists of 325 amino acids, and its N-terminal A-barrel domain is critical for correct folding and function. Outer membrane protein A proteins lacking the C-terminal residues 228 to 325 or residues 194 to 325 are correctly incorporated into the outer membrane and confer all known OmpA phenotypes (41, 42). We also observed that E109, which expresses the truncated OmpA lacking amino acids 301 to 325, has a similar ability to invade into C6 cells to wild-type E44 and E105. In addition to OmpA, type 1 fimbriae and other bacterial components have been implicated in the adhesion and invasion of E. coli to BMECs (3, 7). Expression of type 1 fimbriae is controlled by the inversion of a promotercontaining element upstream of the fim operon (43). In OmpA-deleted mutants, this invertible element is predominantly in the OFF orientation, leading to decreased fimbrial expression. It has been shown that the ompA deletion mutant derived from the fim locked-ON background still exhibited significantly lower adhesion and invasion abilities than the fim locked-ON mutant, suggesting that OmpA is the primary determinant for binding to BMECs (40). Our complementary 688 Ó 2009 American Association of Neuropathologists, Inc. Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 OmpA in E. coli Invasion Into Astrocytes On the other hand, after bacterial infection, C6 cells were still alive and multiplied as efficiently as uninfected control cells up to 7 days in our study (data not shown). The anti-apoptotic factor BclXL is induced after the invasion of E. coli K1 to prevent apoptosis of macrophages (47). In our pilot experiments, the OmpA- strain E91 and MG1655 (OmpA+ , nonpathogenic and noninvasive strain) had approximately 100-fold higher 100% lethal dose compared with the OmpA+ strain E44 (data not shown). Mice died within 48 hours of intracerebral injection of the OmpA+ strains E44, whereas mice challenged with the OmpA- strains E91 or MG1655 survived for 8 days (Fig. 6). The remaining bacteria in the brains increased at 24 and 48 hours postinfection, whereas the bacteria in brains infected with E91 were undetectable at 24 hours postinfection (Table 2). These results indicate that mice survival correlates with bacterial clearance. We suggest intracerebral microglia cells are responsible for the primary clearance of all the bacteria challenged (48). Multiple factors may contribute additively to the difference of clearance of OmpA+ or OmpA- bacteria. For example, E44 was demonstrated to survive in both murine and human macrophage cell lines and in monocytes and macrophages of newborn rats (39). Therefore, E44 might have a better chance of being sustained in microglia. Outer membrane protein AYnegative, but not OmpAYposiive , E. coli might simultaneously activate human neutrophils to produce oxygen radicals (49). Additionally, the OmpA- strain is more susceptible to membrane-acting bactericidal peptides than the wild-type strain (49). Overall, OmpA+ bacteria have all the advantages for intracerebral survival, which may lead to the lethal effects on mouse. Increased GFAP expression is linked to astrogliosis in bacterial meningitis (50). Glial fibrillary acidic protein is important for modulating astrocyte motility and shape by providing structural stability to the astrocyte. We observed that infection of OmpA+ E. coli K1 strains, but not OmpA- E. coli K1 strains or MG1655, induced the expression of GFAP by C6 cells, and that pretreatment with cytochalasin D, LY294002, or genistein inhibited the invasion-induced accumulation of GFAP, suggesting that the bacterial invasive process is a prerequisite for GFAP expression. Thus, we conclude that OmpA is a major determinant of the adherence, invasion, and activation of astrocytes after E. coli infection. Once bacteria infect the CNS, inflammatory molecules such as COX-2, NOS-2, IL-8, and intracellular adhesion molecule 1 are induced, leading to an increase in the permeability of the BBB (23, 51Y53). The inflammatory effects then trigger transendothelial migration of neutrophils that may result in further brain damage. Indeed, we demonstrate that intracerebral E44 infection induced COX-2 and NOS-2 expression, and that COX-2 and NOS-2 colocalized with GFAP in activated astrocytes. Simultaneously, intracerebral injection of E44, but not OmpA- bacteria, also increased neutrophil infiltration in mouse forebrain (Fig. 10). The inflammatory processes progress to include the destruction of the BBB, neutrophil infiltration, neuron damage, and finally, the death of C57BL/6 mice within 48 hours. We also demonstrated that OmpA-mediated binding plays an important role in the early stage of E. coli K1 CNS infection. Preincubation of BMEC with OmpA protein, solubilized from the outer membrane of E44, inhibited the invasion of OmpA+ E. coli (17). Similarly, the purified Nterminal amino acids 1 to 171 of OmpA also decreased the association of E. coli K1 with BMEC in a dose-dependent manner (8). Here, we used the purified recombinant fulllength OmpA protein to protect C6 cells from infection by E44. Furthermore, recombinant OmpA protein protected increased the survival of infected mice up to 80%. Simultaneously, the neutrophil infiltration in brain sections was reduced in the presence of His-OmpA, suggesting that HisOmpA inhibited E44 adhesion to astrocytes, thus preventing their activation and subsequent inflammatory processes. Purified amino-terminal OmpA binds directly to BMECs, whereas a derivative lacking all 4 extracellular loops could not (8). In addition, short peptides corresponding to loops 1 and 2 blocked OmpA+ E. coli invasion of BMECs (17). It remains to be determined which parts of the external loops are required for OmpA-mediated adhesion and invasion of astrocytes. In bacterial meningitis, antibiotic therapy is the first choice for management, but neurologic complications often cannot be averted. Our results that recombinant OmpA protein can prevent the invasion of astrocytes by E. coli and the subsequent inflammation events suggest a new approach for preventing or reducing the neurologic complications that follow astrocyte activation during bacterial meningitis. The possible protective effects and routes of administration of OmpA protein for patients with bacteremia need to be investigated further. Ó 2009 American Association of Neuropathologists, Inc. ACKNOWLEDGMENTS The authors thank K.S. Kim, MD, Division of Pediatric Infectious Diseases, School of Medicine, Johns Hopkins University (Baltimore, MD), for the E. coli strains (E44, E91, E105, E109, E111, and MG1655); Jean-San Chia, MD, Graduate Institute and Department of Microbiology, College of Medicine, National Taiwan University (Taipei, Taiwan), for helpful discussion; and Tim J. Harrison of the Royal Free and University College Medical School (University College London) for critical reading and editing of the manuscript. REFERENCES 1. Shah DK, Daley AJ, Hunt RW, et al. Cerebral white matter injury in the newborn following Escherichia coli meningitis. Eur J Paediatr Neurol 2005;9:13Y17 2. Leib SL, Tauber MG. Pathogenesis of bacterial meningitis. Infect Dis Clin North Am 1999;13:527Y48, vYvi. Review 3. Kim KS. Escherichia coli translocation at the blood-brain barrier. Infect Immun 2001;69:5217Y22 4. Teng CH, Cai M, Shin S, et al. Escherichia coli K1 RS218 interacts with human brain microvascular endothelial cells via type 1 fimbria bacteria in the fimbriated state. Infect Immun 2005;73:2923Y31 5. Hoffman JA, Wass C, Stins MF, et al. The capsule supports survival but not traversal of Escherichia coli K1 across the blood-brain barrier. Infect Immun 1999;67:3566Y70 6. Khan NA, Shin S, Chung JW, et al. Outer membrane protein A and cytotoxic necrotizing factorY1 use diverse signaling mechanisms for Escherichia coli K1 invasion of human brain microvascular endothelial cells. Microb Pathog 2003;35:35Y42 7. Kim KS. Pathogenesis of bacterial meningitis: From bacteraemia to neuronal injury. Nat Rev Neurosci 2003;4:376Y85 689 Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited. Wu et al 8. Shin S, Lu G, Cai M, et al. Escherichia coli outer membrane protein A adheres to human brain microvascular endothelial cells. Biochem Biophys Res Commun 2005;330:1199Y204 9. Koebnik R. Structural and functional roles of the surface-exposed loops of the beta-barrel membrane protein OmpA from Escherichia coli. J Bacteriol 1999;181:3688Y94 10. Klimke WA, Frost LS. Genetic analysis of the role of the transfer gene, traN, of the F and R100-1 plasmids in mating pair stabilization during conjugation. J Bacteriol 1998;180:4036Y43 11. Klimke WA, Rypien CD, Klinger B, et al. The mating pair stabilization protein, TraN, of the F plasmid is an outer-membrane protein with two regions that are important for its function in conjugation. Microbiology 2005;151:3527Y40 12. Schwarz H, Riede I, Sonntag I, et al. Degrees of relatedness of T-even type E. coli phages using different or the same receptors and topology of serologically cross-reacting sites. EMBO J 1983;2:375Y80 13. Morona R, Klose M, Henning U. Escherichia coli K-12 outer membrane protein (OmpA) as a bacteriophage receptor: Analysis of mutant genes expressing altered proteins. J Bacteriol 1984;159:570Y78 14. Riede I, Eschbach ML, Henning U. DNA sequence heterogeneity in the genes of T-even type Escherichia coli phages encoding the receptor recognizing protein of the long tail fibers. Mol Gen Genet 1984;195: 144Y52 15. Montag D, Riede I, Eschbach ML, et al. Receptor-recognizing proteins of T-even type bacteriophages. Constant and hypervariable regions and an unusual case of evolution. J Mol Biol 1987;196:165Y74 16. Pautsch A, Schulz GE. Structure of the outer membrane protein A transmembrane domain. Nat Struct Biol 1998;5:1013Y17 17. Prasadarao NV, Wass CA, Weiser JN, et al. Outer membrane protein A of Escherichia coli contributes to invasion of brain microvascular endothelial cells. Infect Immun 1996;64:146Y53 18. Condorelli DF, Dell’Albani P, Kaczmarek L, et al. Glial fibrillary acidic protein messenger RNA and glutamine synthetase activity after nervous system injury. J Neurosci Res 1990;26:251Y57 19. Chiu FC, Norton WT, Fields KL. The cytoskeleton of primary astrocytes in culture contains actin, glial fibrillary acidic protein, and the fibroblast-type filament protein, vimentin. J Neurochem 1981;37:147Y55 20. Farina C, Aloisi F, Meinl E. Astrocytes are active players in cerebral innate immunity. Trends Immunol 2007;28:138Y45 21. Lieberman AP, Pitha PM, Shin HS, et al. Production of tumor necrosis factor and other cytokines by astrocytes stimulated with lipopolysaccharide or a neurotropic virus. Proc Natl Acad Sci U S A 1989;86:6348Y52 22. Murphy S, Simmons ML, Agullo L, et al. Synthesis of nitric oxide in CNS glial cells. Trends Neurosci 1993;16:323Y28 23. Tsao N, Hsu HP, Lei HY. TNFalpha-induced cyclooxygenase 2 not only increases the vasopermeability of blood-brain barrier but also enhances the neutrophil survival in Escherichia coliYinduced brain inflammation. Prostaglandins Other Lipid Mediat 1999;57:371Y82 24. Azuma I. Review: Inducer of cytokines in vivo: Overview of field and romurtide experience. Int J Immunopharmacol 1992;14:487Y96 25. Dong Y, Benveniste EN. Immune function of astrocytes. Glia 2001;36: 180Y90 26. Salazar-Mather TP, Hokeness KL. Calling in the troops: Regulation of inflammatory cell trafficking through innate cytokine/chemokine networks. Viral Immunol 2003;16:291Y306 27. Kronfeld I, Zsukerman A, Kazimirsky G, et al. Staurosporine induces astrocytic phenotypes and differential expression of specific PKC isoforms in C6 glial cells. J Neurochem 1995;65:1505Y14 28. Mangoura D, Sakellaridis N, Jones J, et al. Early and late passage C-6 glial cell growth: Similarities with primary glial cells in culture. Neurochem Res 1989;14:941Y47 29. Meier C, Oelschlaeger TA, Merkert H, et al. Ability of Escherichia coli isolates that cause meningitis in newborns to invade epithelial and endothelial cells. Infect Immun 1996;64:2391Y99 30. Prasadarao NV, Wass CA, Stins MF, et al. Outer membrane protein AYpromoted actin condensation of brain microvascular endothelial cells is required for Escherichia coli invasion. Infect Immun 1999;67:5775Y83 31. Greiffenberg L, Goebel W, Kim KS, et al. Interaction of Listeria monocytogenes with human brain microvascular endothelial cells: 690 J Neuropathol Exp Neurol Volume 68, Number 6, June 2009 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. InlB-dependent invasion, long-term intracellular growth, and spread from macrophages to endothelial cells. Infect Immun 1998;66:5260Y67 Nizet V, Kim KS, Stins M, et al. Invasion of brain microvascular endothelial cells by group B streptococci. Infect Immun 1997;65:5074Y81 Slevogt H, Seybold J, Tiwari KN, et al. Moraxella catarrhalis is internalized in respiratory epithelial cells by a trigger-like mechanism and initiates a TLR2- and partly NOD1-dependent inflammatory immune response. Cell Microbiol 2007;9:694Y707 Reddy MA, Wass CA, Kim KS, et al. Involvement of focal adhesion kinase in Escherichia coli invasion of human brain microvascular endothelial cells. Infect Immun 2000;68:6423Y30 Lindmo K, Stenmark H. Regulation of membrane traffic by phosphoinositide 3Ykinases. J Cell Sci 2006;119:605Y14 Spring TG, Wold F. The purification and characterization of Escherichia coli enolase. J Biol Chem 1971;246:6797Y802 Torres AG, Kaper JB. Multiple elements controlling adherence of enterohemorrhagic Escherichia coli O157:H7 to HeLa cells. Infect Immun 2003;71:4985Y95 Torres AG, Li Y, Tutt CB, et al. Outer membrane protein A of Escherichia coli O157:H7 stimulates dendritic cell activation. Infect Immun 2006;74:2676Y85 Sukumaran SK, Shimada H, Prasadarao NV. Entry and intracellular replication of Escherichia coli K1 in macrophages require expression of outer membrane protein A. Infect Immun 2003;71:5951Y61 Teng CH, Xie Y, Shin S, et al. Effects of ompA deletion on expression of type 1 fimbriae in Escherichia coli K1 strain RS218 and on the association of E. coli with human brain microvascular endothelial cells. Infect Immun 2006;74:5609Y16 Henning U, Cole ST, Bremer E, et al. Gene fusions using the ompA gene coding for a major outer-membrane protein of Escherichia coli K12. Eur J Biochem 1983;136:233Y40 Bremer E, Cole ST, Hindennach I, et al. Export of a protein into the outer membrane of Escherichia coli K12. Stable incorporation of the OmpA protein requires less than 193 amino-terminal amino-acid residues. Eur J Biochem 1982;122:223Y31 Smith SG, Dorman CJ. Functional analysis of the FimE integrase of Escherichia coli K-12: Isolation of mutant derivatives with altered DNA inversion preferences. Mol Microbiol 1999;34:965Y79 Reddy MA, Prasadarao NV, Wass CA, et al. Phosphatidylinositol 3Ykinase activation and interaction with focal adhesion kinase in Escherichia coli K1 invasion of human brain microvascular endothelial cells. J Biol Chem 2000;275:36769Y74 Ireton K, Payrastre B, Cossart P. The Listeria monocytogenes protein InlB is an agonist of mammalian phosphoinositide 3Ykinase. J Biol Chem 1999;274:17025Y32 Kerr MC, Teasdale RD. Defining macropinocytosis. Traffic 2009;10: 364Y371 Sukumaran SK, Selvaraj SK, Prasadarao NV. Inhibition of apoptosis by Escherichia coli K1 is accompanied by increased expression of BclXL and blockade of mitochondrial cytochrome c release in macrophages. Infect Immun 2004;72:6012Y22 Kreutzberg GW. Microglia: A sensor for pathological events in the CNS. Trends Neurosci 1996;19:312Y18 Fu H, Belaaouaj AA, Dahlgren C, et al. Outer membrane protein A deficient Escherichia coli activates neutrophils to produce superoxide and shows increased susceptibility to antibacterial peptides. Microbes Infect 2003;5:781Y88 Eng LF, Ghirnikar RS, Lee YL. Glial fibrillary acidic protein: GFAP-thirty-one years (1969Y2000). Neurochem Res 2000;25:1439Y51 Kim JM, Oh YK, Lee JH, et al. Induction of proinflammatory mediators requires activation of the TRAF, NIK, IKK and NF-kappaB signal transduction pathway in astrocytes infected with Escherichia coli. Clin Exp Immunol 2005;140:450Y60 Galanakis E, Di Cello F, Paul-Satyaseela M, et al. Escherichia coli K1 induces IL-8 expression in human brain microvascular endothelial cells. Eur Cytokine Netw 2006;17:260Y65 Selvaraj SK, Periandythevar P, Prasadarao NV. Outer membrane protein A of Escherichia coli K1 selectively enhances the expression of intercellular adhesion moleculeY1 in brain microvascular endothelial cells. Microbes Infect 2007;9:547Y57 Ó 2009 American Association of Neuropathologists, Inc. Copyright @ 2009 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
© Copyright 2025 Paperzz