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










Publication year range
1.
Crit Care Med ; 25(1): 101-5, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8989184

ABSTRACT

OBJECTIVE: To compare a recombinant bactericidal/permeability-increasing protein variant and a recombinant endotoxin-neutralizing protein. DESIGN: Randomized, blinded, controlled study, using a rat model of sepsis. SETTING: Animal research facility. SUBJECTS: Male Wistar rats. INTERVENTIONS: An inoculum of 1.5 x 10(7) to 1.8 x 10(8) Escherichia coli O18ac K1, implanted in the peritoneum, produced bacteremia in 95% of animals after 1 hr. One hour after E. coli challenge, animals received recombinant bactericidal/permeability-increasing protein variant, recombinant endotoxin-neutralizing protein, or saline intravenously, followed by ceftriaxone and gentamicin intramuscularly. MEASUREMENTS AND MAIN RESULTS: Twenty-four (85.7%) of 28 animals receiving recombinant endotoxin-neutralizing protein (p < .001 vs. control) survived 7 days compared with nine (33.3%) of 27 recombinant bactericidal/permeability-increasing protein variant-treated (p < .001 vs. control) and two (6.5%) of 31 control animals. CONCLUSIONS: Both recombinant endotoxin-neutralizing protein and recombinant bactericidal/permeability-increasing protein variant improved survival. Recombinant endotoxin-neutralizing protein was superior to recombinant bactericidal/permeability-increasing protein variant in its protective effect at the doses tested. Our results suggest that both proteins may be useful in the treatment of human Gram-negative sepsis.


Subject(s)
Anti-Infective Agents/therapeutic use , Escherichia coli Infections/therapy , Gram-Negative Bacterial Infections/therapy , Sepsis/therapy , Animals , Anti-Infective Agents/pharmacology , Ceftriaxone/therapeutic use , Disease Models, Animal , Drug Therapy, Combination , Escherichia coli Infections/mortality , Gentamicins/therapeutic use , Male , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Sepsis/microbiology , Sepsis/mortality , Survival Analysis
2.
Arch Surg ; 131(2): 200-5; discussion 206, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8611079

ABSTRACT

BACKGROUND: Bactericidal/permeability-increasing protein (BPI) binds lipopolysaccharide and neutralizes its toxic effects in vitro and in endotoxemic animals. Our recent work identified physiologically significant interactions between BPI, lipopolysaccharide, and mononuclear cells. OBJECTIVE: To determine whether the interaction between BPI and mononuclear cells is receptor mediated. DESIGN: Labeled BPI was incubated with THP-1 cells in the presence of up to 100-fold excess of unlabeled BPI. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis and Western blotting were performed to evaluate competitive binding and total uptake of BPI. Crosslinking was performed to determine whether BPI binds to a single protein entity. Acid washing experiments and flow cytometric analysis were performed to determine whether BPI remains on the cellular surface. Finally, flow cytometry analysis was used to determine whether BPI incubation with THP-1 cells affects the surface expression of the lipopolysaccharide-binding protein-lipopolysaccharide receptor CD14. RESULTS: Labeled BPI uptake was not inhibited by the presence of 100-fold excess of unlabeled BPI at 37 degrees C or 4 degrees C in the presence of azide. Uptake was not saturable under either condition with incubation concentrations up to 10 microgram/mL. Cross-linking did not show BPI bound to a single entity. Acid washing and flow cytometry experiments disclosed rapid internalization of BPI. Finally, BPI uptake by THP-1 cells had no effect on the surface expression of CD14. CONCLUSIONS: Bactericidal/permeability-increasing protein is rapidly internalized by mononuclear cells in a nonspecific fashion not saturable at very high doses, which is consistent with pinocytosis. This process may represent a disposal mechanism for lipopolysaccharide in closed-space infections and may be partially responsible for the rapid clearance of BPI from the peripheral circulation.


Subject(s)
Anti-Infective Agents/metabolism , Blood Bactericidal Activity , Blood Proteins/metabolism , Leukemia, Monocytic, Acute/metabolism , Membrane Proteins , Pinocytosis , Antimicrobial Cationic Peptides , Binding, Competitive , Blotting, Western , Cell Line , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukocytes, Mononuclear/metabolism , Lipopolysaccharide Receptors/drug effects , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/metabolism , Protein Binding , Receptors, Cell Surface/metabolism , Receptors, Drug/metabolism , Sodium Dodecyl Sulfate
3.
Nephrol Dial Transplant ; 11(1): 101-8, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8649615

ABSTRACT

BACKGROUND: Previous studies on the permeability of cellulosic and synthetic dialysers for bacterial-derived cytokine-inducing substances gave conflicting results. We tried to study this issue as close to the in-vivo situation as possible. METHODS: An in-vitro dialysis circuit with whole human blood present in the blood compartment of cuprophane (Cup), polysulphone (PS), and polyamide (PA) dialysers was employed; sterile filtrates derived from Pseudomonas aeruginosa cultures were added to the dialysate. We studied the induction of interleukin-1 beta (IL-1 beta) by plasma samples taken from the blood compartment as well as the induction of IL-1 beta and interleukin-1 receptor antagonist (IL-1Ra) in mononuclear cells separated from whole blood after circulation by radioimmunoassay and polymerase chain reaction. RESULTS: Plasma samples from the blood side of all dialysers induced IL-1 beta from non-circulated mononuclear cells after addition of pseudomonas filtrates to the dialysate; the maximal amount of IL-1 beta induced by samples from the blood compartment was 4.8 +/- 1.2 ng/ml for Cup, 1.9 +/- 0.5 ng/ml for PS, and 2.0 +/- 0.6 ng/ml for PA. Mononuclear cells separated after contaminated dialysis will all types of dialysers expressed increased mRNA levels for IL-1 beta and IL-1Ra. Production of IL-1Ra by cells separated after contaminated dialysis was determined after Cup and PS dialysis; there was increased production of IL-1Ra by these cells (Cup, 10.3 +/- 4.2; PS, 7.3 +/- 2.5 ng/ml) compared to cells separated after sterile dialysis (Cup, 5.6 +/- 2.1, P < 0.05; PS, 4.5 +/- 1.1 ng/ml, n.s.) or from non-circulated blood (Cup experiments, 4.7 +/- 1.5, P < 0.05; PS experiments, 4.1 +/- 1.2 ng/ml, n.s.). CONCLUSIONS: These data suggest penetration of cytokine-inducing substances through both cellulosic and synthetic dialysers. Differences between dialysers may exist regarding extent and time course of penetration. The detection of cytokine mRNA as well as the measurement of IL-1Ra synthesis is a more sensitive marker for the transfer of cytokine-inducing substances through dialyser membranes than the measurement of IL-1 beta protein synthesis.


Subject(s)
Dialysis Solutions/metabolism , Interleukin-1/biosynthesis , Leukocytes, Mononuclear/metabolism , Membranes, Artificial , Renal Dialysis/instrumentation , Sialoglycoproteins/biosynthesis , Base Sequence , Humans , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/genetics , Leukocytes, Mononuclear/drug effects , Lipopolysaccharides/pharmacology , Molecular Sequence Data , Polymerase Chain Reaction , Pseudomonas aeruginosa , RNA, Messenger/metabolism , Radioimmunoassay , Sialoglycoproteins/genetics
4.
Antimicrob Agents Chemother ; 39(12): 2813-5, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8593028

ABSTRACT

A chimeric protein consisting of the N-terminal domain of lipopolysaccharide-binding protein and the C-terminal domain of bactericidal/permeability-increasing protein demonstrated a dose-dependent survival benefit (P = 0.001) and reduced endotoxin levels (P < 0.01) in neutropenic rats with Pseudomonas aeruginosa sepsis. This lipopolysaccharide-binding protein-bactericidal/ permeability-increasing peptide has favorable pharmacokinetics and antiendotoxin properties which may be of value for human sepsis.


Subject(s)
Acute-Phase Proteins , Blood Proteins/therapeutic use , Carrier Proteins/therapeutic use , Membrane Glycoproteins , Membrane Proteins , Pseudomonas Infections/drug therapy , Sepsis/drug therapy , Animals , Antimicrobial Cationic Peptides , Blood Proteins/pharmacokinetics , Carrier Proteins/pharmacokinetics , Cell Membrane/metabolism , Colony Count, Microbial , Endotoxins/analysis , Female , Limulus Test , Neutropenia/complications , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Sepsis/metabolism , Sepsis/microbiology
5.
J Clin Invest ; 95(4): 1947-52, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7706502

ABSTRACT

Bactericidal/permeability-increasing protein (BPI) is a neutrophil primary granule protein that inhibits effects of LPS in vitro. The current study examined the effects of BPI on hemodynamics, mortality, and circulating endotoxin and cytokines in conscious rats with endotoxic shock. Catheters were implanted into the right femoral artery and vein. 1 d later, human recombinant BPI (10 mg/kg) or vehicle was intravenously injected immediately, 30 min, or 2 h after intravenous injection of LPS (7.5 mg/kg). Mean arterial pressure (MAP) and heart rate were monitored and blood was collected before and after injection. BPI given immediately or 30 min after LPS prevented the LPS-induced reduction in MAP at 4-8 h and markedly reduced mortality. BPI given 2 h after LPS injection had no protective effect. BPI treated immediately after LPS reduced the circulating levels of endotoxin and IL-6 but increased the circulating levels of TNF. We propose that BPI exerts its protective effect through a TNF-independent mechanism, by inhibiting endotoxin-stimulated production of IL-6.


Subject(s)
Anti-Infective Agents/therapeutic use , Blood Proteins/therapeutic use , Membrane Proteins , Shock, Septic/therapy , Animals , Antimicrobial Cationic Peptides , Blood Pressure/drug effects , Endotoxins/blood , Endotoxins/pharmacology , Heart Rate/drug effects , Hemodynamics/drug effects , Hypotension/drug therapy , Interleukin-6/blood , Lipopolysaccharides/pharmacology , Male , Rats , Rats, Sprague-Dawley , Recombinant Proteins/therapeutic use , Shock, Septic/mortality , Time Factors , Tumor Necrosis Factor-alpha/analysis
6.
Infect Immun ; 62(9): 3930-6, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8063410

ABSTRACT

Bactericidal/permeability-increasing protein (BPI), a human neutrophil granule protein, has been shown to bind lipopolysaccharide (LPS) and neutralize LPS-mediated cytokine production in adherent monocytes and the whole-blood system. In this study we investigated the ability of recombinant human BPI (rBPI) to inhibit LPS-induced vascular endothelial cell (EC) injury and activation. rBPI inhibited significantly both rough and smooth LPS-mediated injury for cultured bovine brain microvessel ECs, as measured by lactic dehydrogenase release, and blocked the LPS-induced interleukin-6 (IL-6) release from human umbilical vein ECs in a dose-dependent manner. BPI was able to inhibit LPS-mediated EC injury or activation whether it was added before or at the same time with LPS, but delaying the time of addition of rBPI resulted only in a partial inhibition. BPI also inhibited LPS-induced tumor necrosis factor alpha, IL-1 beta, and IL-6 release from human whole blood. This inhibition of tumor necrosis factor alpha, IL-1 beta, and IL-6 release from whole blood was maximal when BPI was premixed with LPS before addition to blood and was partial when BPI was added simultaneously with LPS, but no inhibition was observed when the addition of rBPI was delayed for 5 min. These findings suggest that rBPI is a potent inhibitor of LPS-mediated responses in ECs and whole blood and underscore the potential use of BPI in treatment or prevention of endotoxic shock. In contrast, the anti-lipid A monoclonal antibodies HA-1A and E5 were ineffective in inhibiting LPS-mediated EC injury and activation as well as LPS-induced cytokine release in whole blood.


Subject(s)
Blood Bactericidal Activity , Blood Proteins/pharmacology , Endothelium, Vascular/drug effects , Lipopolysaccharides/toxicity , Membrane Proteins , Animals , Antibodies, Monoclonal/immunology , Antimicrobial Cationic Peptides , Cattle , Cells, Cultured , Humans , Interleukin-1/metabolism , Interleukin-6/metabolism , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/metabolism
7.
Lancet ; 344(8920): 429-31, 1994 Aug 13.
Article in English | MEDLINE | ID: mdl-7520106

ABSTRACT

Endotoxin initiates the systemic inflammatory response, haemodynamic changes, and multi-organ failure that may occur as a consequence of systemic gram-negative bacterial infection. The serum protein lipopolysaccharide-binding protein (LBP) binds to the lipid A component of bacterial endotoxin and facilitates its delivery to the CD14 antigen on the macrophage, where inflammatory cytokines are released and a cascade of host mediators is initiated. The neutrophil granular protein bactericidal/permeability-increasing protein (BPI) competes with LBP for endotoxin binding and functions as a molecular antagonist of LBP-endotoxin interactions. We have measured concentrations of both proteins in body fluids from 49 consecutive patients. In 16 of 17 samples of fluid from closed-space infections, BPI was present in greater concentration than LBP (median BPI/LBP ratio 7.6 [95% CI 2.32-22.1]). The ratio of BPI and LBP was not significantly different from 1.0 in abdominal fluid from 10 patients with peritonitis (ratio 0.235 [0.18-0.47]), whereas the BPI/LBP ratio was low in 22 non-infected body fluids (0.01 [0.001-0.04]) and concentrations of both proteins approached those in normal human plasma. BPI concentrations were directly correlated with the quantity of neutrophils within clinical samples (rs = 0.81, p < 0.0001). Thus, within abscess cavities BPI is available in sufficient quantities for effective competition with LBP for endotoxin. BPI may attenuate the local inflammatory response and the systemic toxicity of endotoxin release during gram-negative infections.


Subject(s)
Abscess/pathology , Acute-Phase Proteins , Bacterial Infections/pathology , Blood Bactericidal Activity , Blood Proteins/analysis , Body Fluids/chemistry , Carrier Proteins/analysis , Membrane Glycoproteins , Membrane Proteins , Neutrophils , Peritonitis/pathology , Abscess/immunology , Abscess/microbiology , Aged , Antimicrobial Cationic Peptides , Bacterial Infections/immunology , Bacterial Infections/microbiology , Binding, Competitive , Blood Proteins/chemistry , Blood Proteins/immunology , Carrier Proteins/chemistry , Carrier Proteins/immunology , Female , Humans , Inflammation , Male , Middle Aged , Peritonitis/immunology , Peritonitis/microbiology , Permeability
8.
Ann Surg ; 220(1): 77-85, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8024362

ABSTRACT

OBJECTIVE: The in vivo neutralizing activities of an anti-lipopolysaccharide (LPS) antibody HA-1A (Centoxin [Centocor, Malvern, PA]), a human immunoglobulin M monoclonal antibody, and of bactericidal/permeability-increasing protein (BPI), an endogenously produced human LPS-neutralizing protein, were studied in a primate model of lethal Escherichia coli bacteremia. SUMMARY BACKGROUND DATA: HA-1A has been used with variable success against LPS activity in some animal models and in a recently reported clinical trial. However, no data assessing the efficacy of this agent in subhuman primates is available. Bactericidal/permeability-increasing protein is a product of polymorphomononuclear cells (PMNs) that is stored in azurophilic granules and exhibits LPS-neutralizing activity in vitro and in some in vivo models. METHODS: Immediately after E. coli infusion and in a blinded fashion, three baboons were treated with BPI (5 mg/kg bolus infusion and 95 micrograms/kg/min infusion over 4 hr). Three animals received 3 mg/kg BW of HA-1A, whereas another three baboons received a placebo treatment. RESULTS: The BPI-treated animals demonstrated significantly (p < 0.03) lower circulating LPS-limulus amoebocyte lysate (LAL) activity compared with the control animals, but this reduction in LPS-LAL activity was not associated with improved survival. HA-1A treatment did not reduce LPS-LAL activity. However, both BPI and HA-1A treatment did attenuate the pro-inflammatory cytokine response. CONCLUSION: The current data suggests that incomplete neutralization of endotoxin activity does not alter mortality from severe bacteremia. Given the diversity of mediator production under such circumstances, a strategy of combination therapy in the form of anti-lipopolysaccharide and anticytokine treatment may be necessary to achieve optimal survival.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bacteremia/therapy , Blood Proteins/therapeutic use , Endotoxins/therapeutic use , Escherichia coli Infections/therapy , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Bacteremia/immunology , Bacteremia/metabolism , Bacteremia/microbiology , Bacteremia/physiopathology , Blood Proteins/pharmacology , Endotoxins/immunology , Endotoxins/pharmacology , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/physiopathology , Hemodynamics/drug effects , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/metabolism , Interleukin-6/metabolism , Limulus Test , Lipopolysaccharides/blood , Molecular Sequence Data , Papio , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Sialoglycoproteins/drug effects , Sialoglycoproteins/metabolism , Survival Rate , Time Factors , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
9.
Crit Care Med ; 22(4): 553-8, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8143463

ABSTRACT

OBJECTIVES: To study the toxicology and pharmacology of the endotoxin-neutralizing agent, bactericidal/permeability-increasing protein. DESIGN: Prospective, randomized, placebo-controlled laboratory study. SETTING: Academic research laboratory. SUBJECTS: CD-1 mice (n = 259); Sprague Dawley rats (n = 26); New Zealand White rabbits (n = 19). INTERVENTIONS: Pharmacokinetics of intravenously injected bactericidal/permeability-increasing protein was assessed in mice. Toxicology was tested in mice and rats. Efficacy of intravenously administered bactericidal/permeability-increasing protein as an endotoxin-neutralizing agent was tested in mice, rats, and rabbits. MEASUREMENTS AND MAIN RESULTS: Administration of a single 10-mg/kg bolus injection of bactericidal/permeability-increasing protein resulted in no alterations in hematologic, renal, or hepatic function, activity level, or weight gain in animals observed over a 7-day study period. A single bolus injection (10 mg/kg) of bactericidal/permeability-increasing protein protected 15 of 16 mice from a lethal endotoxin challenge (mortality rate 1/16 [6.25%]) compared with a 100% (16/16) mortality rate in the saline-treated controls (p < .001). Bactericidal/permeability-increasing protein administered up to 1 hr after endotoxin provided significant protection against lethal endotoxin challenge. Furthermore, bactericidal/permeability-increasing protein reduced the induration and dermal necrosis observed in the localized dermal Shwartzman reaction. CONCLUSIONS: Bactericidal/permeability-increasing protein is a potent antiendotoxin that neutralizes endotoxin in vivo and prevents mortality in animal models of lethal endotoxemia.


Subject(s)
Blood Proteins/administration & dosage , Membrane Proteins , Neutrophils/drug effects , Shock, Septic/drug therapy , Animals , Antimicrobial Cationic Peptides , Blood Proteins/pharmacokinetics , Blood Proteins/toxicity , Endotoxins/antagonists & inhibitors , Female , Humans , Injections, Intravenous , Lipid A/metabolism , Mice , Rabbits , Random Allocation , Rats , Rats, Sprague-Dawley , Shock, Septic/metabolism , Shock, Septic/mortality
10.
Crit Care Med ; 22(4): 559-65, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8143464

ABSTRACT

OBJECTIVE: To compare the endotoxin-binding and -neutralizing properties of bactericidal/permeability-increasing protein, the human monoclonal antiendotoxin antibody HA-1A, and the murine antiendotoxin antibody E5. DESIGN: Prospective, randomized, placebo-controlled laboratory study. SETTING: Biotechnology company research laboratory. SUBJECTS: Female CD-1 mice. INTERVENTIONS: Recombinant bactericidal/permeability-increasing protein, HA-1A, a human immunoglobulin M monoclonal antibody raised against Escherichia coli J5 (Rc) endotoxin, and E5, a murine immunoglobulin M monoclonal antibody raised against E. coli J5 endotoxin, were compared in the following assays: a) binding to rough lipopolysaccharide immobilized onto microtiter plates; b) inhibition of lipopolysaccharide activity in the limulus amebocyte lysate assay; c) inhibition of lipopolysaccharide-induced cytokine release in whole blood; and d) protection against lethal endotoxin challenge in CD-1 mice. MEASUREMENTS AND MAIN RESULTS: The binding affinity of bactericidal/permeability-increasing protein for immobilized lipopolysaccharide is apparently greater than the binding affinity of HA-1A or E5. Bactericidal/permeability-increasing protein neutralized lipopolysaccharide activity in the chromogenic limulus amebocyte lysate assay, while neither monoclonal antibody inhibited lipopolysaccharide activity. Similarly, bactericidal/permeability-increasing protein reduced lipopolysaccharide-mediated tumor necrosis factor production in human whole blood in vitro, whereas monoclonal antibodies had slight (HA-1A) or no (E5) effect on lipopolysaccharide activity in this system. Administration of bactericidal/permeability-increasing protein gave > 90% protection against an LD60 dose of endotoxin in CD-1 mice, while treatment with HA-1A or E5 did not improve survival rate. CONCLUSIONS: Neither monoclonal antibody was as effective as bactericidal/permeability-increasing protein at binding or neutralizing endotoxin in vitro or in vivo. The potent endotoxin-binding and -neutralizing properties of bactericidal/permeability-increasing protein indicate that it might be useful in the treatment of endotoxin-related disorders in humans.


Subject(s)
Antibodies, Monoclonal/metabolism , Blood Proteins/metabolism , Endotoxins/immunology , Membrane Proteins , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antimicrobial Cationic Peptides , Blood Proteins/immunology , Female , Humans , Lethal Dose 50 , Lipopolysaccharides/metabolism , Mice , Prospective Studies , Shock, Septic/metabolism
11.
J Clin Immunol ; 14(2): 120-33, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8195314

ABSTRACT

Human neutrophil azurophilic granules contain an approximately 55-kDa protein, known as bactericidal/permeability-increasing protein (BPI), which possesses a high-affinity binding domain for the lipid A component of lipopolysaccharide (LPS). The in vivo LPS neutralizing activity of exogenous BPI was studied in a model of lethal Escherichia coli bacteremia. Five baboons were treated with BPI (5 mg/kg bolus injection followed by a 95 micrograms/kg/min BPI infusion over 4 hr), while four additional animals received a genetically engineered variant of BPI (NCY103). Five animals received a placebo treatment and served as controls. Both wild-type rhBPI and NCY103 significantly (P < 0.05) decreased blood levels of LPS throughout an 8-hr evaluation period following live bacterial challenge. Two hours following E. coli administration, LPS levels peaked in the controls, at 6.86 +/- 3.22 ng/ml, whereas LPS levels were 3.39 +/- 2.1 ng/ml in the BPI group and 2.04 +/- 1.18 ng/ml in the NCY103 group. Tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 levels likewise were attenuated in the treatment groups, whereas circulating sTNFR I was significantly (P < 0.05) reduced only in the BPI group. Leukocytopenia and granulocytopenia were significantly (P < 0.02) lessened in the BPI group, by an average of 59% leukocytopenia and 65% granulocytopenia, respectively. This study supports the concept of E. coli LPS neutralization by BPI in vivo and demonstrates that a moderate (70%) reduction in peak LPS-LAL activity is sufficient to alter some hematologic and cytokine manifestations of bacteremia.


Subject(s)
Anti-Infective Agents/therapeutic use , Bacteremia/therapy , Blood Proteins/therapeutic use , Escherichia coli Infections/therapy , Membrane Proteins , Shock, Septic/therapy , Animals , Antimicrobial Cationic Peptides , Interleukin-6/biosynthesis , Neutrophils/immunology , Papio , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Proteins/therapeutic use , Tumor Necrosis Factor-alpha/biosynthesis
12.
Arch Surg ; 129(2): 220-6, 1994 Feb.
Article in English | MEDLINE | ID: mdl-7508221

ABSTRACT

OBJECTIVE: To evaluate changes in levels of polymorphonuclear leukocyte surface bactericidal/permeability-increasing protein (BPI), plasma BPI, and plasma lipopolysaccharide (LPS) binding protein (LBP) in normal human volunteers administered Escherichia coli LPS and in patients with sepsis and gram-negative infections. DESIGN: Survey; case series. SETTING: Clinical research center and surgical intensive care unit of a medical school and an associated tertiary care hospital. PATIENTS OR OTHER PARTICIPANTS: Volunteers (n = 10) screened prior to study by history and physical examination to exclude those with underlying diseases or hematologic abnormalities. Consecutive sample of surgical intensive care unit patients (n = 10) meeting criteria for sepsis syndrome with gram-negative infection. An additional patient with systemic inflammatory response syndrome but no gram-negative infection. All patients were studied on meeting the criteria. Three of the patients with sepsis syndrome and the patient with systemic inflammatory response syndrome were evaluated on recovery (approximately 25 days after initial study). Because these studies in volunteers and patients overlapped temporally, the control values were those of volunteers evaluated prior to LPS administration. No matching was employed. MEASUREMENTS AND RESULTS: Compared with controls, LPS-challenged volunteers and patients with sepsis both exhibited significant granulocytosis (P < .01) and increased concentrations of polymorphonuclear leukocyte surface BPI (P < .01) and of plasma LBP (P < .01). Plasma BPI concentrations were increased (P < .01) in volunteers following LPS administration. There was a trend toward increased concentrations of plasma BPI in patients, but this was not significant relative to controls. Maximum concentrations of plasma LBP were approximately 250- and 3000-fold higher than plasma BPI concentrations in endotoxemic volunteers and in patients, respectively. CONCLUSIONS: Circulating polymorphonuclear leukocytes increase expression of BPI in response to LPS or gram-negative sepsis. Subsequently, concentrations of plasma BPI and LBP increase. Because both LBP and BPI bind to LPS, it is suggested that endogenously derived plasma levels of BPI are likely to be inadequate to compete for LPS binding to the much more abundant LBP in the circulation.


Subject(s)
Acute-Phase Proteins/analysis , Blood Proteins/analysis , Blood Proteins/chemistry , Carrier Proteins/blood , Endotoxins/blood , Escherichia coli , Gram-Negative Bacterial Infections/blood , Membrane Glycoproteins , Membrane Proteins , Neutrophils/chemistry , Adult , Antimicrobial Cationic Peptides , Blood Bactericidal Activity , Endotoxins/adverse effects , Humans , Leukocyte Count , Lipopolysaccharides/adverse effects , Lipopolysaccharides/blood , Male , Neutrophils/pathology , Prospective Studies , Survival Rate , Syndrome , Time Factors
13.
Clin Nephrol ; 40(6): 346-51, 1993 Dec.
Article in English | MEDLINE | ID: mdl-7507806

ABSTRACT

Several proteins modify the biological response to lipopolysaccharide (LPS). Both bactericidal/permeability-increasing factor (BPI), a protein stored in neutrophils, and the acute phase protein LPS-binding protein (LBP) bind to LPS; however, BPI inhibits while LBP enhances binding of LPS to leukocytes and subsequent induction of cytokines. We investigated plasma levels of BPI, LBP, elastase and C5a before, during and after hemodialysis (HD). Six patients were dialysed with Cuprophane (Cup) and polysulfone (PS) low-flux dialyzers on two consecutive HD sessions. There was a significant, 10.9 +/- 2.8-fold increase in BPI after 4-hour HD compared to predialysis and a 4.4 +/- 1.6-fold increase in elastase after 4-hour HD using Cup. Plasma levels of BPI and elastase decreased rapidly after the dialysis session. HD with PS resulted in a smaller, but still significant rise in BPI (3.7 +/- 1.6-fold at 4 hours) and elastase (1.69 +/- 0.2-fold at 4 hours). Levels for BPI and elastase were similar in the arterial and venous blood lines of the dialyzer. Plasma levels of LBP did not change during or after the HD session. These data indicate that BPI, but not LBP is released during HD with Cup and to a lesser extent with PS. Activation of neutrophils and release of BPI during HD may influence the biological response to bacterial products possibly introduced during HD.


Subject(s)
Acute-Phase Proteins/analysis , Blood Bactericidal Activity , Blood Proteins/analysis , Carrier Proteins/blood , Kidney Failure, Chronic/therapy , Membrane Glycoproteins , Membrane Proteins , Renal Dialysis , Antimicrobial Cationic Peptides , Cellulose/analogs & derivatives , Complement C5a/analysis , Enzyme-Linked Immunosorbent Assay , Humans , Immunoassay , Kidney Failure, Chronic/blood , Membranes, Artificial , Pancreatic Elastase/blood , Polymers , Sulfones
14.
J Immunol ; 151(8): 4258-65, 1993 Oct 15.
Article in English | MEDLINE | ID: mdl-8409400

ABSTRACT

Serum proteins play an important role in LPS-induced cell activation. The LPS binding protein (LBP) enhances cellular responses to LPS, whereas the polymorphonuclear leukocyte product bactericidal/permeability-increasing protein (BPI) inhibits LPS-induced cell activation. In this study the influences of LBP and BPI, two proteins with opposite effects, but with considerable sequence homology, on LPS-induced mononuclear phagocytic cell cytokine release was studied. LBP was shown to enhance LPS-induced TNF-alpha, IL-6, and IL-8 release by mononuclear phagocytic cells, whereas BPI inhibited the release of these cytokines. Furthermore, the effects of LBP and BPI on LPS-induced cytokine release by mononuclear phagocytic cells were shown to be counteractive. BPI interfered with the enhancing effect of LBP on the LPS-induced cytokine release. At high LBP to BPI ratios, BPI could no longer inhibit LBP-induced enhancement. In accordance, increasing concentrations of BPI abrogated the LBP effect. Next, it was shown that LBP and BPI compete for binding to LPS by using an assay system that detects binding of free BPI to an anti-BPI mAb. LPS prevented binding of BPI to anti-BPI mAb, whereas preincubation of LPS with LBP prevented the LPS-induced inhibition. Also, it was observed that both BPI and LBP inhibited LPS activity in the chromogenic LAL assay. We conclude from this study that LBP and BPI have counteractive effects on LPS-induced mononuclear phagocytic cell cytokine release by competing for binding to LPS.


Subject(s)
Acute-Phase Proteins , Blood Proteins/pharmacology , Carrier Proteins/pharmacology , Cytokines/metabolism , Lipopolysaccharides/antagonists & inhibitors , Membrane Glycoproteins , Membrane Proteins , Phagocytes/drug effects , Antimicrobial Cationic Peptides , Binding, Competitive , Blood Proteins/metabolism , Carrier Proteins/metabolism , Cell Line , Humans , Limulus Test , Lipopolysaccharides/metabolism , Phagocytes/metabolism
15.
Blood Purif ; 11(2): 134-40, 1993.
Article in English | MEDLINE | ID: mdl-7506040

ABSTRACT

Bactericidal/permeability-increasing protein (BPI) is a natural constituent of human neutrophils. Recombinant BPI has been shown to bind to bacterial lipopolysaccharide (LPS), and to neutralize the ability of LPS to stimulate inflammatory cells in vitro and in vivo. BPI shares sequence homology and immunocrossreactivity with another endogenous LPS binding protein, lipopolysaccharide binding protein (LBP). Despite the homology, these proteins have opposite effects on LPS. LBP mediates cell activation by low, otherwise nonstimulatory concentrations, while BPI neutralizes LPS bioactivity. Exogenous LPS binding proteins in the form of monoclonal antibodies have been developed with the goal of generating antiendotoxin therapeutics to treat gram-negative sepsis and related syndromes. Here we show that LPS-binding and neutralizing properties of BPI compare favorably with two monoclonal antibodies tested, HA-1A and XMMEN-OE5. BPI also competes effectively with LBP for LPS. Thus, BPI may represent an endogenous LPS-regulatory molecule suitable for use as a potent antiendotoxin therapeutic.


Subject(s)
Blood Bactericidal Activity , Blood Proteins/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Membrane Proteins , Neutrophils , Receptors, Immunologic/drug effects , Animals , Antimicrobial Cationic Peptides , Female , Humans , Lipopolysaccharide Receptors , Mice , Permeability , Recombinant Proteins/pharmacology
16.
J Immunol ; 148(2): 532-7, 1992 Jan 15.
Article in English | MEDLINE | ID: mdl-1729370

ABSTRACT

Systemic release of endotoxin (LPS) after Gram-negative infection initiates a cascade of host cytokines that are thought to be the direct cause of shock, multisystem organ failure, and death. Endogenous LPS-binding proteins may play a role in regulating LPS toxicity in vivo. The human neutrophil granule protein bactericidal/permeability-increasing protein (BPI) shares sequence homology and immunocrossreactivity with an acute phase lipopolysaccharide binding protein (LBP) which has been shown to bind to LPS and accelerate LPS activation of neutrophils and macrophages. Although structurally similar, LBP and BPI are apparently functionally antagonistic. We previously showed that BPI inhibits LPS-mediated neutrophil activation in vitro. Here we demonstrate that BPI binds to LPS near the lipid A domain, and formation of the LPS-BPI complex abrogates detrimental host responses to LPS. For example, BPI blocks LPS-stimulated TNF release in vitro and in vivo, and LPS complexed to BPI is not pyrogenic in rabbits. Results demonstrating that BPI is released by stimulated human neutrophils further support the idea that BPI functions extracellularly in vivo to neutralize endotoxin. Taken together, these data argue that BPI neutralizes the toxic effects of LPS in vivo, and that BPI may represent a new therapeutic approach to the treatment of endotoxic shock.


Subject(s)
Blood Bactericidal Activity , Blood Proteins/pharmacology , Lipopolysaccharides/toxicity , Membrane Proteins , Neutrophils/immunology , Animals , Antimicrobial Cationic Peptides , Blood Proteins/metabolism , Fever/chemically induced , Fever/prevention & control , Humans , In Vitro Techniques , Mice , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Permeability , Rabbits , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
17.
Blood ; 76(4): 825-34, 1990 Aug 15.
Article in English | MEDLINE | ID: mdl-2200540

ABSTRACT

The ontogeny of a 57-Kd cationic antimicrobial protein (CAP57) that has substantial similarities to bactericidal permeability increasing protein (BPI) has been determined immunocytochemically. CAP57 was detected in the granules of mature peripheral blood neutrophils. However, it was absent from other cells of the peripheral blood: eosinophils, red blood cells (RBCs), and mononuclear cells. In human bone marrow, CAP57 was confined to the neutrophilic series. The earliest stage of development of the myeloid cells at which CAP57 was demonstrated was the promyelocyte. Double immunofluorescent labeling showed that CAP57 was detected in cells positive for myeloperoxidase. The absence of lactoferrin in certain cells (promyelocytes) containing CAP57 indicated that CAP57 was synthesized and packaged in a population of granules prior to the development of granules that contain lactoferrin. CAP57 could not be demonstrated in HL60 cells either by enzyme-linked immunosorbent assay (ELISA) or by immunocytochemistry. However, the presence of another granule-associated cationic antimicrobial protein of molecular weight 37 Kd (CAP37) was readily detected in undifferentiated HL60 cells. Amino acid sequence analysis showed that CAP57 and BPI were identical. Further indication of the identity between CAP57 and BPI was that monoclonal anti-CAP57 antibodies cross reacted with BPI. Sucrose density-gradient centrifugations showed CAP57 was confined to a granule population that exhibited a buoyant density intermediate of the previously described light and heavy azurophil granules. Further resolution of the individual azurophil granule populations by Percoll density-gradient centrifugation revealed that CAP57 was most concentrated in the density range of 1.093 to 1.100 g/cc. These results strongly suggest the unique finding that CAP57 may be associated with a heretofore unreported granule type.


Subject(s)
Blood Proteins/metabolism , Membrane Proteins , Neutrophils/metabolism , Amino Acid Sequence , Amino Acids/analysis , Antibodies, Monoclonal/immunology , Antimicrobial Cationic Peptides , Blood Proteins/analysis , Blood Proteins/immunology , Bone Marrow/metabolism , Bone Marrow/ultrastructure , Bone Marrow Cells , Cell Line , Centrifugation, Density Gradient , Cross Reactions/immunology , Cytoplasmic Granules/metabolism , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Leukemia/blood , Leukemia/pathology , Molecular Sequence Data , Neutrophils/cytology
18.
J Immunol ; 144(2): 662-6, 1990 Jan 15.
Article in English | MEDLINE | ID: mdl-2295804

ABSTRACT

Neutrophil granules contain proteins important in host defense against bacterial pathogens. Granule proteins released from activated neutrophils facilitate opsonization, phagocytosis, tissue digestion, and antimicrobial activity. Three similar, if not identical, neutrophil proteins, bactericidal/permeability-increasing protein (BPI), 57,000 m.w. cationic antimicrobial protein, and bactericidal protein have been described that specifically kill gram negative bacteria. Since LPS is a structure common to all gram-negative bacteria, we investigated whether the microbicidal protein BPI affects biologic activity of LPS in vitro. Human neutrophils can be activated both in vitro and in vivo by LPS. Upon stimulation, surface expression of CR1 and CR3 increases markedly. Using flow microfluorimetry, we analyzed surface expression of CR1 and CR3 as a measure of neutrophil stimulation in response to LPS. CR up-regulation on neutrophils was TNF independent, suggesting direct LPS stimulation of neutrophils in this system. Purified BPI completely inhibited CR up-regulation on neutrophils stimulated with both rough and smooth LPS chemotypes at 1.8 to 3.6 nM (100 to 200 ng/ml). By comparison, the polypeptide antibiotic polymyxin B completely inhibited the same dose of LPS at 0.4 nM. The inhibitory activity of BPI appeared to be specific for LPS because neutrophil stimulation by formylated peptide or TNF was unaffected. The specificity of BPI for LPS was further demonstrated by inhibition of LPS activity in the limulus amebocyte lysate assay. Therefore, the role of BPI in infection may not be limited to its microbicidal activity, but it may also regulate the neutrophil response to LPS.


Subject(s)
Blood Proteins/pharmacology , Endotoxins/antagonists & inhibitors , Membrane Proteins , Neutrophils/physiology , Antimicrobial Cationic Peptides , Blood Bactericidal Activity , Cell Membrane Permeability , Humans , In Vitro Techniques , Kinetics , Limulus Test , Lipid A/antagonists & inhibitors , Molecular Weight , Receptors, Complement/metabolism , Tumor Necrosis Factor-alpha/pharmacology
19.
Proc Natl Acad Sci U S A ; 86(14): 5610-4, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2501794

ABSTRACT

Nine polypeptide peaks with antibiotic activity were resolved from human polymorphonuclear leukocyte azurophil granule membranes. All but 1 of the 12 constituent polypeptides were identified by N-terminal sequence analysis. Near quantitative recovery of protein and activity permitted an assessment of the contribution of each species to the overall respiratory-burst-independent antimicrobial capacity of the cell. Three uncharacterized polypeptides were discovered, including two broad-spectrum antibiotics. One of these, a defensin that we have designated human neutrophil antimicrobial peptide 4, was more potent than previously described defensins but represented less than 1% of the total protein. The other, named azurocidin, was abundant and comparable to bactericidal permeability-increasing factor in its contribution to the killing of Escherichia coli.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Blood Proteins/isolation & purification , Neutrophils/analysis , Amino Acid Sequence , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/pharmacology , Candida albicans/drug effects , Chromatography, High Pressure Liquid , Enterococcus faecalis/drug effects , Escherichia coli/drug effects , Humans , Microbial Sensitivity Tests , Molecular Sequence Data , Molecular Weight
20.
J Biol Chem ; 264(19): 11200-3, 1989 Jul 05.
Article in English | MEDLINE | ID: mdl-2500436

ABSTRACT

Primary (azurophil) granules of neutrophils contain proteins which play a major role in the killing and digestion of bacteria in the phagolysosome. We have isolated and characterized a novel antimicrobial peptide from the azurophil granule fraction of discontinuous Percoll gradients. We have named this peptide human neutrophil peptide 4 (HNP-4) based on its structural similarity to a group of antimicrobial polypeptides known as defensins (HNP 1-3). Using size exclusion and reverse-phase high performance liquid chromatography, HNP-4 was purified to homogeneity as judged by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and amino-terminal sequence analysis. The amino acid sequence determined from isolated HNP-4 and from tryptic fragments of reduced and alkylated peptide is: NH2-Val-Cys-Ser-Cys-Arg-Leu-Val-Phe-Cys-Arg-Arg-Thr-Glu- Leu-Arg-Val-Gly-Asn-Cys-Leu-Ile-Gly-Gly-Val-Ser-Phe-Thr-Tyr-Cys-Cys-Thr- Arg-Val - COOH. Based on this sequence, HNP-4 has a calculated molecular weight of 3715 and a theoretical pI of 8.61. HNP-4 shows structural similarity to the family of three human defensins. HNP-4 and the defensins have identical cysteine backbones and, like the defensins, HNP-4 is rich in arginine (15.2 mol %). However, the amino acids at 22 of the 33 positions differ between HNP-4 and human defensins. Further, HNP-4 is significantly more hydrophobic than the defensins, as determined by its retention time on reverse-phase high performance liquid chromatography. In vitro, purified HNP-4 was shown to kill Escherichia coli, Streptococcus faecalis, and Candida albicans. Compared to a mixture of the other human defensins, HNP-4 was found to be approximately 100 times more potent against E. coli and four times more potent against both S. faecalis and C. albicans.


Subject(s)
Blood Proteins/isolation & purification , Neutrophils/analysis , alpha-Defensins , Amino Acid Sequence , Amino Acids/analysis , Blood Bactericidal Activity , Blood Proteins/physiology , Candida albicans , Chromatography, Gel , Chromatography, High Pressure Liquid , Cytoplasmic Granules/analysis , Electrophoresis, Polyacrylamide Gel , Enterococcus faecalis , Escherichia coli , Humans , Isoelectric Point , Molecular Sequence Data , Molecular Weight , Neutrophils/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...