Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Infect Immun ; 68(6): 3349-51, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10816484

ABSTRACT

A difficulty that has emerged in the development and preclinical evaluation of adjuvant therapies for gram-negative sepsis is the lack of easily studied animal models that closely mimic human infection. An objective of this study was to adapt a previously described model of infection in burned mice to rats with a defined bacterial strain of Escherichia coli. Challenge with two colonies of live E. coli O18:K1:H7 bacteria into an 8% full-thickness burn of the dorsal skin surface of rats produced predictable bacteremia at 24 to 48 h and 80 to 100% mortality at 3 to 4 days. E. coli O18:K1:H7 was approximately 10-million-fold more virulent than several other gram-negative bacterial strains. The model should be a useful tool in studying the pathogenicity of burn wound infections and in evaluating the efficacy of novel adjuvant therapies for gram-negative sepsis.


Subject(s)
Bacteremia/microbiology , Burns/complications , Disease Models, Animal , Escherichia coli Infections/microbiology , Gram-Negative Bacterial Infections/microbiology , Wound Infection/microbiology , Animals , Bacteremia/mortality , Burns/mortality , Escherichia coli Infections/mortality , Gram-Negative Bacterial Infections/mortality , Male , Rats , Rats, Sprague-Dawley , Wound Infection/mortality
2.
Infect Immun ; 68(5): 2566-72, 2000 May.
Article in English | MEDLINE | ID: mdl-10768945

ABSTRACT

Complexes containing lipopolysaccharide (LPS) and three outer membrane proteins (OMPs) are released by gram-negative bacteria incubated in human serum and into the circulation in an experimental model of sepsis. The same OMPs are bound by immunoglobulin G (IgG) in the cross-protective antiserum raised to Escherichia coli J5 (anti-J5 IgG). This study was performed to identify the three OMPs. The 35-kDa OMP was identified as outer membrane protein A (OmpA) by immunoblotting studies using OmpA-deficient bacteria and recombinant OmpA protein. The 18-kDa OMP was identified as peptidoglycan-associated lipoprotein (PAL) based on peptide sequences from the purified protein and immunoblotting studies using PAL-deficient bacteria. The 5- to 9-kDa OMP was identified as murein lipoprotein (MLP) based on immunoblotting studies using MLP-deficient bacteria. The studies identify the OMPs released into human serum and into the circulation in an experimental model of sepsis as OmpA, PAL, and MLP.


Subject(s)
Bacterial Outer Membrane Proteins/analysis , Escherichia coli/chemistry , Lipoproteins/analysis , Peptidoglycan/analysis , Proteoglycans , Animals , Bacterial Outer Membrane Proteins/immunology , Escherichia coli/metabolism , Escherichia coli Proteins , Humans , Lipoproteins/immunology , Peptidoglycan/immunology , Rabbits
3.
J Infect Dis ; 181(3): 1034-43, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10720528

ABSTRACT

Prior studies indicate that 3 bacterial outer-membrane proteins (OMPs) are released into serum associated with lipopolysaccharide (LPS) and are bound by IgG in antiserum to Escherichia coli J5 (anti-J5 IgG). The present studies analyzed the interaction of the OMPs with anti-J5 IgG and evaluated their release in an infected burn model of gram-negative sepsis. Affinity purification studies were performed on filtrates of bacteria incubated in human serum and plasma from rats with sepsis by use of O chain-specific anti-LPS IgG and anti-J5 IgG. All 3 OMPs were captured from septic rat blood by anti-LPS IgG. Release of OMPs into serum was highest for immature bacterial cultures and was increased by antibiotics in vitro and in vivo. Anti-J5 IgG selectively captured an 18-kDa OMP released into serum and into plasma from septic rats. The results raise the possibility that anti-J5 IgG may, in part, protect via anti-OMP antibodies.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Escherichia coli/immunology , Gram-Negative Bacterial Infections/metabolism , Immune Sera/metabolism , Immunoglobulin G/metabolism , Sepsis/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/immunology , Bacterial Outer Membrane Proteins/blood , Bacterial Outer Membrane Proteins/isolation & purification , Humans , Mice , Rabbits , Rats
4.
J Infect Dis ; 176(5): 1260-8, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9359727

ABSTRACT

The binding of IgG in antiserum to Escherichia coli J5 to the surface of Enterobacteriaceae and to cell wall fragments released from serum-exposed bacteria was studied in a search for potentially protective epitopes other than lipopolysaccharide (LPS). IgG titers to multiple heterologous gram-negative smooth bacteria increased following incubation of the bacteria in serum and decreased following absorption with serum-exposed heterologous bacteria. IgG eluted from absorbing bacteria bound to at least three conserved bacterial outer membrane proteins (OMPs), but not LPS, as assessed by immunoblotting. The same OMPs were present in LPS-containing macromolecular cell wall fragments released by incubation of heterologous gram-negative bacteria in human serum. Part of the protection offered by J5 antiserum could be from binding of IgG to conserved OMPs at the bacterial surface or to OMPs in cell-wall fragments released from dying bacteria.


Subject(s)
Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Escherichia coli/immunology , Immune Sera/immunology , Animals , Gram-Negative Bacteria/immunology , Humans , Mice , Rabbits
5.
J Infect Dis ; 176(5): 1313-21, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9359733

ABSTRACT

Distributions of immunoreactive interleukin-1 (IL-1) and lipopolysaccharide (LPS) were studied in the tissues of rats after intravenous injection of purified LPS or live Escherichia coli bacteria. IL-1 staining in the spleen peaked at 4-8 h, colocalized with LPS in marginal zone macrophages, and was undetectable 24 h after injection, whereas LPS staining peaked at 24 h and was detectable for 4 weeks. The tissue IL-1 response was similar for LPS and live bacteria. Thus, tissue IL-1 is down-regulated within hours despite maintenance of LPS in the same cells for weeks. Macrophages in liver and lung had only slight IL-1 staining despite intense staining for LPS. Tissue IL-1 production appears to be differentially regulated after gram-negative bacteremia; LPS cleared by liver and lung macrophages elicit minimal IL-1, whereas there is high local IL-1 production in the marginal zone of the spleen that may increase immune responses to bacterial wall antigens.


Subject(s)
Bacteremia/immunology , Endotoxemia/immunology , Interleukin-1/biosynthesis , Lipopolysaccharides/metabolism , Animals , Interleukin-1/analysis , Lipopolysaccharides/analysis , Liver/immunology , Lung/immunology , Macrophages/immunology , Male , Rats , Rats, Sprague-Dawley , Spleen/immunology
6.
Infect Immun ; 65(6): 2160-7, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9169746

ABSTRACT

The 18-kDa cationic protein CAP18 is an antimicrobial protein isolated from rabbit granulocytes that binds lipopolysaccharide (LPS) and inhibits many of its biological activities. We covalently coupled a synthetic peptide representing amino acids 106 to 138 of CAP18 to human immunoglobulin G (IgG) by using the heterobifunctional linker N-succinimidyl-3-(2-pyridyidithio)propionate. The ability of CAP18(106-138)-IgG to bind and neutralize LPS in whole blood in the presence and absence of anticoagulants was studied. Both CAP18(106-138) and CAP18(106-138)-IgG significantly suppressed LPS-induced tumor necrosis factor (TNF) production in whole blood in the absence of anticoagulants. EDTA potentiated the ability of CAP18(106-138) and CAP18(106-138)-IgG to decrease LPS-induced TNF production in a dose-dependent manner. In contrast, heparin inhibited the ability of CAP18(106-138) and CAP18(106-138)-IgG to suppress LPS-induced TNF production. EDTA also enhanced LPS capture in a fluid-phase binding assay that utilizes magnetic anti-IgG beads to capture CAP18(106-138)-IgG (and bound [3H]LPS) in whole blood. In contrast, heparin inhibited the binding dose dependently. We conclude that CAP18(106-138)-IgG binds to and neutralizes LPS in whole blood in the absence of anticoagulants. Further studies of its protective efficacy in animal models are warranted. Caution should be used in interpreting assays that measure the binding and neutralization of LPS in whole blood in the presence of calcium-binding anticoagulants or heparin.


Subject(s)
Anticoagulants/pharmacology , Antimicrobial Cationic Peptides , Carrier Proteins/pharmacology , Immunoglobulin G/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Animals , Carrier Proteins/metabolism , Cathelicidins , Humans , Immunoglobulin G/metabolism , Lipopolysaccharides/blood , Lipopolysaccharides/metabolism , Rabbits , Tumor Necrosis Factor-alpha/biosynthesis
7.
J Infect Dis ; 175(3): 621-32, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9041334

ABSTRACT

Although type-specific IgG directed to the O-polysaccharide antigen of bacterial lipopolysaccharide (LPS) is protective in most models of LPS or bacterial challenge, no currently available IgG binds to LPS from all gram-negative bacteria. The ability of a peptide-IgG conjugate, CAP18(106-138)-IgG, to bind and neutralize LPS, to kill gram-negative bacteria, and to protect in a sensitized mouse model of LPS toxicity was studied. CAP18(106-138)-IgG bound LPS from multiple gram-negative bacteria in four different binding assays. In a fluid-phase RIA, half-maximal binding of 5 microg/mL 3H-labeled LPS occurred at 5-10 microg/mL CAP18(106-138)-IgG, similar to binding with monoclonal type-specific IgG. CAP18(106-138)-IgG neutralized LPS, as assessed by LPS-induced coagulation of limulus amebocyte lysate and production of tumor necrosis factor in vitro, was bactericidal for a wide range of gram-negative bacteria, and decreased LPS-induced lethality in sensitized mice. Antibacterial peptide-IgG conjugates merit further study as a novel adjunctive therapy for gram-negative sepsis.


Subject(s)
Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides , Carrier Proteins/chemistry , Gram-Negative Bacteria/drug effects , Lipopolysaccharides/chemistry , Animals , Anti-Bacterial Agents/pharmacology , Binding Sites , Binding, Competitive , Cathelicidins , Cells, Cultured , Immunoglobulin G/chemistry , Limulus Test , Mice , Protein Binding , Tumor Necrosis Factor-alpha/biosynthesis
8.
Mol Med ; 2(3): 334-48, 1996 May.
Article in English | MEDLINE | ID: mdl-8784786

ABSTRACT

BACKGROUND: Understanding the processes that control selective eosinophilia is of fundamental importance in a variety of human diseases (e.g., allergies, parasitic infections, malignancy). Interleukin 5, an eosinophil-specific growth and activating factor, and eotaxin appear to collaborate in this process. Eotaxin is a recently described chemotactic factor that belongs to the C-C (or beta) chemokine family and has been implicated in animal and human eosinophilic inflammatory states. We have recently reported the molecular characterization of murine eotaxin and now report the biological properties of purified recombinant murine eotaxin in vitro and in vivo in the presence or absence of interleukin 5 (IL-5) in mice. MATERIALS AND METHODS: Murine eotaxin was expressed in bacteria and purified by affinity chromatography and HPLC. Activity was tested in vitro by examining chemotactic and calcium flux responses of purified murine leukocytes. Additionally, desensitization of calcium flux responses to other chemokines, eosinophil survival assays, and basophil histamine release were examined. Finally, eotaxin was delivered to wild-type or IL-5 transgenic mice and the host response was examined. RESULTS: Eotaxin had activity only when the recombinant molecule had the native mature amino terminus and contained the first 25 amino acids of the mature protein. It was active in vitro at an effective concentration between 10 and 100 ng/ml in both chemotaxis and calcium flux assays toward eosinophils, but not macrophages or neutrophils. Furthermore, intranasal or subcutaneous application of eotaxin selectively recruited large numbers of eosinophils into the mouse lung and skin, respectively, only in the presence of interleukin 5. Macrophage inflammatory protein-1 alpha, a related C-C chemokine active on eosinophils, and eotaxin were not able to cross-desensitize. Eotaxin had no affect on the in vitro survival of eosinophils and did not induce basophil histamine release. CONCLUSIONS: Mouse eotaxin is an eosinophil specific chemoattractant that has a markedly enhanced effect in vivo in the presence of another eosinophil selective cytokine IL-5, and utilizes a signal transduction receptor pathway that is distinct from that utilized by macrophage inflammatory protein-1 alpha. This data suggests that the development of tissue eosinophilia in vivo involves a two-step mechanism elicited by interleukin 5 and eotaxin.


Subject(s)
Basophils/immunology , Calcium/blood , Chemokines, CC , Chemotactic Factors, Eosinophil/pharmacology , Chemotaxis, Leukocyte/drug effects , Eosinophilia/immunology , Eosinophils/physiology , Interleukin-5/physiology , Lung Diseases/immunology , Receptors, Chemokine , Receptors, Cytokine/physiology , Animals , Basophils/drug effects , Cell Survival/drug effects , Cells, Cultured , Chemokine CCL11 , Cloning, Molecular , Cytokines/isolation & purification , Cytokines/pharmacology , Eosinophilia/blood , Eosinophilia/chemically induced , Eosinophils/cytology , Eosinophils/drug effects , Escherichia coli , Factor Xa/metabolism , Histamine Release/drug effects , Humans , Interleukin-5/biosynthesis , Interleukin-5/genetics , Lung Diseases/blood , Lung Diseases/chemically induced , Macrophages/drug effects , Macrophages/physiology , Mice , Mice, Inbred CBA , Mice, Inbred Strains , Mice, Transgenic , Receptors, CCR3 , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
9.
J Exp Med ; 177(1): 89-97, 1993 Jan 01.
Article in English | MEDLINE | ID: mdl-8418211

ABSTRACT

The use of monoclonal antibodies (mAbs) directed to lipid A for the therapy of gram-negative sepsis is controversial. In an attempt to understand their biologic basis of action, we used a fluid-phase radioimmunoassay to measure binding between bacterial lipopolysaccharide (LPS) and two IgM mAbs directed to lipid A that are being evaluated for the treatment of gram-negative bacterial sepsis. Both antibodies bound 3H-LPS prepared from multiple strains of gram-negative bacteria when large excesses of antibody were used, although binding was modest and only slightly greater than control preparations. We also studied the ability of each anti-lipid A antibody to neutralize some of the biological effects of LPS in vitro. Despite large molar excesses, neither antibody neutralized LPS as assessed by the limulus lysate test, by a mitogenic assay for murine splenocytes, or by the production of cytokines interleukin (IL)-1, IL-6, or tumor necrosis factor from human monocytes in culture medium or in whole blood. Our experiments do not support the hypothesis that either of these anti-lipid A mAbs function by neutralizing the toxic effects of LPS.


Subject(s)
Antibodies, Monoclonal/immunology , Lipid A/immunology , Lipopolysaccharides/immunology , Adult , Animals , Antibodies, Monoclonal, Humanized , Cytokines/biosynthesis , Humans , Lipopolysaccharides/metabolism , Mice , Mice, Inbred BALB C , Monocytes/metabolism , Neutralization Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...