Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
Add more filters










Publication year range
1.
Immunol Rev ; 313(1): 358-375, 2023 01.
Article in English | MEDLINE | ID: mdl-36161656

ABSTRACT

C3 is a key complement protein, located at the nexus of all complement activation pathways. Extracellular, tissue, cell-derived, and intracellular C3 plays critical roles in the immune response that is dysregulated in many diseases, making it an attractive therapeutic target. However, challenges such as very high concentration in blood, increased acute expression, and the elevated risk of infections have historically posed significant challenges in the development of C3-targeted therapeutics. This is further complicated because C3 activation fragments and their receptors trigger a complex network of downstream effects; therefore, a clear understanding of these is needed to provide context for a better understanding of the mechanism of action (MoA) of C3 inhibitors, such as pegcetacoplan. Because of C3's differential upstream position to C5 in the complement cascade, there are mechanistic differences between pegcetacoplan and eculizumab that determine their efficacy in patients with paroxysmal nocturnal hemoglobinuria. In this review, we compare the MoA of pegcetacoplan and eculizumab in paroxysmal nocturnal hemoglobinuria and discuss the complement-mediated disease that might be amenable to C3 inhibition. We further discuss the current state and outlook for C3-targeted therapeutics and provide our perspective on which diseases might be the next success stories in the C3 therapeutics journey.


Subject(s)
Hemoglobinuria, Paroxysmal , Humans , Hemoglobinuria, Paroxysmal/drug therapy , Complement C3/metabolism , Complement C3/pharmacology , Complement Activation , Complement C5/pharmacology , Complement C5/therapeutic use , Complement Inactivating Agents/pharmacology , Complement Inactivating Agents/therapeutic use
2.
Br J Pharmacol ; 180(4): 422-440, 2023 02.
Article in English | MEDLINE | ID: mdl-36251578

ABSTRACT

BACKGROUND AND PURPOSE: Traumatic haemorrhage (TH) is the leading cause of potentially preventable deaths that occur during the prehospital phase of care. No effective pharmacological therapeutics are available for critical TH patients yet. Here, we identify terminal complement activation (TCA) as a therapeutic target in combat casualties and evaluate the efficacy of a TCA inhibitor (nomacopan) on organ damage and survival in vivo. EXPERIMENTAL APPROACH: Complement activation products and cytokines were analysed in plasma from 54 combat casualties. The correlations between activated complement pathway(s) and the clinical outcomes in trauma patients were assessed. Nomacopan was administered to rats subjected to lethal TH (blast injury and haemorrhagic shock). Effects of nomacopan on TH were determined using survival rate, organ damage, physiological parameters, and laboratory profiles. KEY RESULTS: Early TCA was associated with systemic inflammatory responses and clinical outcomes in this trauma cohort. Lethal TH in the untreated rats induced early TCA that correlated with the severity of tissue damage and mortality. The addition of nomacopan to a damage-control resuscitation (DCR) protocol significantly inhibited TCA, decreased local and systemic inflammatory responses, improved haemodynamics and metabolism, attenuated tissue and organ damage, and increased survival. CONCLUSION AND IMPLICATIONS: Previous findings of our and other groups revealed that early TCA represents a rational therapeutic target for trauma patients. Nomacopan as a pro-survival and organ-protective drug, could emerge as a promising adjunct to DCR that may significantly reduce the morbidity and mortality in severe TH patients while awaiting transport to critical care facilities.


Subject(s)
Complement C5 , Shock, Hemorrhagic , Rats , Animals , Complement C5/pharmacology , Shock, Hemorrhagic/drug therapy , Complement Activation , Immunologic Factors/pharmacology , Phenotype
3.
Immunol Rev ; 313(1): 376-401, 2023 01.
Article in English | MEDLINE | ID: mdl-36398537

ABSTRACT

Over a century after the discovery of the complement system, the first complement therapeutic was approved for the treatment of paroxysmal nocturnal hemoglobinuria (PNH). It was a long-acting monoclonal antibody (aka 5G1-1, 5G1.1, h5G1.1, and now known as eculizumab) that targets C5, specifically preventing the generation of C5a, a potent anaphylatoxin, and C5b, the first step in the eventual formation of membrane attack complex. The enormous clinical and financial success of eculizumab across four diseases (PNH, atypical hemolytic uremic syndrome (aHUS), myasthenia gravis (MG), and anti-aquaporin-4 (AQP4) antibody-positive neuromyelitis optica spectrum disorder (NMOSD)) has fueled a surge in complement therapeutics, especially targeting diseases with an underlying complement pathophysiology for which anti-C5 therapy is ineffective. Intensive research has also uncovered challenges that arise from C5 blockade. For example, PNH patients can still face extravascular hemolysis or pharmacodynamic breakthrough of complement suppression during complement-amplifying conditions. These "side" effects of a stoichiometric inhibitor like eculizumab were unexpected and are incompatible with some of our accepted knowledge of the complement cascade. And they are not unique to C5 inhibition. Indeed, "exceptions" to the rules of complement biology abound and have led to unprecedented and surprising insights. In this review, we will describe initial, present and future aspects of protein inhibitors of the complement cascade, highlighting unexpected findings that are redefining some of the mechanistic foundations upon which the complement cascade is organized.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Hemoglobinuria, Paroxysmal , Humans , Complement System Proteins/metabolism , Complement Activation , Hemoglobinuria, Paroxysmal/drug therapy , Atypical Hemolytic Uremic Syndrome/drug therapy , Complement C5/metabolism , Complement C5/pharmacology , Complement C5/therapeutic use , Complement Inactivating Agents/therapeutic use , Complement Inactivating Agents/pharmacology
4.
Cells ; 11(7)2022 03 28.
Article in English | MEDLINE | ID: mdl-35406699

ABSTRACT

INTRODUCTION: Both increased activity of the complement system (CS) and the role of the pituitary hormone prolactin (PRL) are implicated in osteoarthritis (OA) pathogenesis. Besides, Cathepsin D (CatD) activity is increased in the context of OA and can exert not only proteolytic but also non-proteolytic effects on cells. For the first time, possible crosstalk between two separate humoral systems: the CS and the PRL hormone systems in chondrocytes are examined together. METHODS: Primary human articular chondrocytes (hAC) were stimulated with complement protein C5 (10 µg /mL), PRL (25 ng/mL), CatD (100 ng/mL), or anaphylatoxin C5a (25 ng/mL) for 24 h or 72 h, while unstimulated cells served as controls. In addition, co-stimulations of C5 or PRL with CatD were carried out under the same conditions. The influence of the stimulants on cell viability, cell proliferation, and metabolic activity of hAC, the chondrosarcoma cell line OUMS-27, and endothelial cells of the human umbilical cord vein (HUVEC) was investigated. Gene expression analysis of C5a receptor (C5aR1), C5, complement regulatory protein CD59, PRL, PRL receptor (PRLR), CatD, and matrix metal-loproteinases (MMP)-13 were performed using real-time PCR. Also, collagen type (Col) I, Col II, C5aR1, CD59, and PRL were detected on protein level using immunofluorescence labeling. RESULTS: The stimulation of the hAC showed no significant impairment of the cell viability. C5, C5a, and PRL induced cell growth in OUMS-27 and HUVEC, but not in chondrocytes. CatD, as well as C5, significantly reduced the gene expression of CatD, C5aR1, C5, and CD59. PRLR gene expression was likewise impaired by C5, C5a, and PRL+CatD stimulation. On the protein level, CatD, as well as C5a, decreased Col II as well as C5aR1 synthesis. CONCLUSIONS: The significant suppression of the C5 gene expression under the influence of PRL+CatD and that of CD59 via PRL+/-CatD and conversely a suppression of the PRLR gene expression via C5 alone or C5a stimulation indicates an interrelation between the two mentioned systems. In addition, CatD and C5, in contrast to PRL, directly mediate possible negative feedback of their own gene expression.


Subject(s)
Chondrocytes , Osteoarthritis , Cathepsin D/metabolism , Chondrocytes/metabolism , Complement C5/metabolism , Complement C5/pharmacology , Complement C5a/pharmacology , Complement System Proteins/metabolism , Endothelial Cells/metabolism , Humans , Osteoarthritis/metabolism , Prolactin/metabolism , Prolactin/pharmacology
5.
Amino Acids ; 53(1): 143-147, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33398524

ABSTRACT

The complement component C5 inhibitory peptide zilucoplan is currently in phase III clinical trials for myasthenia gravis (MG). Despite being at an advanced stage of clinical development, there have been no published reports in the literature detailing its chemical synthesis. In this work, we describe an approach for the chemical synthesis of zilucoplan and validate that the synthesised compound blocks LPS-induced C5a production from human blood.


Subject(s)
Complement C5/antagonists & inhibitors , Complement Inactivating Agents/chemical synthesis , Peptides, Cyclic/chemical synthesis , Complement C5/chemical synthesis , Complement C5/chemistry , Complement C5/pharmacology , Complement Inactivating Agents/chemistry , Complement Inactivating Agents/pharmacology , Humans , Inhibitory Concentration 50 , Lipopolysaccharides/pharmacology , Molecular Structure , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Solid-Phase Synthesis Techniques
6.
Osteoarthritis Cartilage ; 21(12): 1958-67, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24041966

ABSTRACT

OBJECTIVE: Tissue-based xenografts such as cartilage are rejected within weeks by humoral and cellular mechanisms that preclude its clinical application in regenerative medicine. The problem could be overcome by identifying key molecules triggering rejection and the development of genetic-engineering strategies to counteract them. Accordingly, high expression of α1,2-fucosyltransferase (HT) in xenogeneic cartilage reduces the galactose α1,3-galactose (Gal) antigen and delays rejection. Yet, the role of complement activation in this setting is unknown. DESIGN: To determine its contribution, we assessed the effect of inhibiting C5 complement component in α1,3-galactosyltransferase-knockout (Gal KO) mice transplanted with porcine cartilage and studied the effect of human complement on porcine articular chondrocytes (PAC). RESULTS: Treatment with an anti-mouse C5 blocking antibody for 5 weeks enhanced graft survival by reducing cellular rejection. Moreover, PAC were highly resistant to complement-mediated lysis and primarily responded to human complement by releasing IL-6 and IL-8. This occurred even in the absence of anti-Gal antibody and was mediated by both C5a and C5b-9. Indeed, C5a directly triggered IL-6 and IL-8 secretion and up-regulated expression of swine leukocyte antigen I (SLA-I) and adhesion molecules on chondrocytes, all processes that enhance cellular rejection. Finally, the use of anti-human C5/C5a antibodies and/or recombinant expression of human complement regulatory molecule CD59 (hCD59) conferred protection in correspondence with their specific functions. CONCLUSIONS: Our study demonstrates that complement activation contributes to rejection of xenogeneic cartilage and provides valuable information for selecting approaches for complement inhibition.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cartilage, Articular/drug effects , Chondrocytes/transplantation , Complement C5/antagonists & inhibitors , Complement C5a/immunology , Complement Membrane Attack Complex/immunology , Graft Survival/drug effects , Heterografts/immunology , Transplantation, Heterologous/methods , Animals , CD59 Antigens/immunology , Cartilage, Articular/cytology , Complement C5/immunology , Complement C5/pharmacology , Galactosyltransferases/genetics , Graft Rejection/prevention & control , Histocompatibility Antigens Class I , Histocompatibility Antigens Class II/immunology , Humans , Interleukin-6/immunology , Interleukin-8/immunology , Mice , Mice, Knockout , Swine
7.
Blood ; 99(6): 2207-13, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11877299

ABSTRACT

Neutrophils are continuously released from the bone marrow (BM), and this release is accelerated during inflammation. This study compared the mechanical properties of mature neutrophils within the BM and the circulating blood, as well as the role of microtubule rearrangement in the release of neutrophils from the BM in rats. Neutrophils isolated from the BM were stiffer than neutrophils in the circulating blood, using magnetic twisting cytometry. BM neutrophils also contained more F-actin within the submembrane region than circulating neutrophils when examined using confocal microscopy, suggesting that mature quiescent neutrophils within the BM are stiffer than circulating neutrophils because of increased formation of F-actin beneath the plasma membrane. Complement protein 5 fragments or formylmethionyl-leucylphenylalanine (fMLP) induced a stiffening response within 2 minutes that was greater in circulating than in BM neutrophils. This stiffening required F-actin formation within the submembrane region but not microtubule rearrangement in both circulating and BM neutrophils. fMLP-induced shape changes were more pronounced in circulating than in BM neutrophils, which showed fewer and smaller pseudopods and fewer membrane irregularities. In vivo, fMLP induced neutropenia, sequestration of neutrophils within the pulmonary capillaries, and release of neutrophils from the BM. Studies using colchicine demonstrated that rearrangement of microtubules was not required for any of these processes but was required for normal trafficking of neutrophils through the pulmonary capillaries.


Subject(s)
Inflammation Mediators/pharmacology , Neutrophils/drug effects , Neutrophils/physiology , Actins/analysis , Actins/biosynthesis , Actins/physiology , Animals , Biomechanical Phenomena , Blood Cells/drug effects , Blood Cells/physiology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Size/drug effects , Complement C5/administration & dosage , Complement C5/pharmacology , Inflammation Mediators/administration & dosage , Male , Microtubules/metabolism , Microtubules/physiology , Microtubules/ultrastructure , N-Formylmethionine Leucyl-Phenylalanine/administration & dosage , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Pliability , Rats , Rats, Inbred Lew
8.
Surg Infect (Larchmt) ; 2(4): 303-10, 2001.
Article in English | MEDLINE | ID: mdl-12593706

ABSTRACT

BACKGROUND: In vitro studies suggest that polymorphonuclear neutrophils (PMN) can damage endothelial cells (EC) by releasing hydrogen peroxide. In vivo this can lead to anasarca secondary to capillary leakage of fluid, protein, and electrolytes. The result is multiple organ dysfunction syndrome, which is associated with high mortality. In vivo, circulating PMN-EC interactions take place in the presence of plasma, and we have shown previously that plasma affords protection to EC from PMN-mediated damage. METHODS: Human umbilical vein endothelial cells were primed with cytokines, cultured to a confluent monolayer, and coincubated with normal human PMNs. Cytotoxicity was assayed by gamma scintigraphy, plasma C5 was determined by sepharose column elution, and H(2)O(2) was assayed by R-Phycoerythrin fluorescence. RESULTS: Addition of C5, but not C3, to RPMI resulted in EC cytoprotection equivalent to adding whole serum. Removal of C5 from serum using F(ab')(2) rabbit IgG anti-human C5 coupled to CNBr-activated 4 sepharose beads resulted in significant loss of EC cytoprotection against H(2)O(2)-mediated damage, whereas adding back C5 restored the cytoprotection. C5 also reduced H(2)O(2)-mediated destruction of R-Phycoerythrin. CONCLUSIONS: The data suggest that the protection of EC against hydrogen peroxide-mediated damage is partly mediated through complement component C5.


Subject(s)
Complement C5/immunology , Complement C5/pharmacology , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/injuries , Hydrogen Peroxide/adverse effects , Hydrogen Peroxide/immunology , Neutrophils/immunology , Oxidants/adverse effects , Oxidants/immunology , Plasma/immunology , Animals , Complement Activation/drug effects , Complement Activation/immunology , Endothelium, Vascular/drug effects , Female , Humans , In Vitro Techniques , Male , Neutrophils/drug effects , Rabbits , Umbilical Veins/drug effects , Umbilical Veins/immunology , Umbilical Veins/injuries
9.
J Immunol ; 165(4): 2183-9, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10925305

ABSTRACT

Anaphylatoxin derived from the fifth complement component (C5a) in the presence of IL-3 induces continuous leukotriene C4 generation and IL-4 and IL-13 expression in human basophils for a period of 16-18 h. This indicates that the G protein-coupled C5a receptor (C5aR) can induce long-lasting cellular responses. Using anti-N-terminal C5aR Abs, C-terminal C5a hexapeptide analogs, and pertussis toxin, we demonstrate that the putative activation site of the C5aR is both necessary and sufficient for these late cellular responses. Furthermore, continuous pertussis toxin-sensitive G protein-coupled receptor activation and receptor-ligand interaction is ongoing and required during the entire period of product release. However, the late basophil responses have a more stringent requirement for optimal receptor activation. Leukotriene C4 generation appears to be influenced mostly by the way the receptor is activated, because the most active hexapeptide is a superagonist for this response. By contrast, C5adesarg, lacking the C-terminal arginine, induces minimal lipid mediator formation but is fully active to induce IL-4 production and is even a superagonist for IL-13 release. Nevertheless, IL-4/IL-13 synthesis in response to C5adesarg could be blocked by both C-terminal antagonistic peptide as well as anti-N-terminal C5aR Abs, indicating only minor differences of ligand-receptor interactions between C5a and C5adesarg. Taken together, our data demonstrate that long-lasting and continuous signaling occurs through a limited activation domain of the C5aR, which can differentially promote separate basophil functions.


Subject(s)
Antigens, CD/physiology , Basophils/immunology , Basophils/metabolism , Complement C5a/metabolism , Interleukin-13/biosynthesis , Interleukin-4/biosynthesis , Leukotriene C4/blood , Receptors, Complement/physiology , Cells, Cultured , Complement Activation , Complement C5/immunology , Complement C5/pharmacology , Complement C5a/physiology , Complement C5a, des-Arginine/metabolism , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Dose-Response Relationship, Immunologic , Humans , Interleukin-13/blood , Interleukin-4/blood , Leukotriene C4/antagonists & inhibitors , Leukotriene C4/biosynthesis , Peptide Fragments/physiology , Receptor, Anaphylatoxin C5a , Receptors, Complement/antagonists & inhibitors , Time Factors
10.
J Am Soc Nephrol ; 10(1): 128-35, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9890318

ABSTRACT

The objective of the study was to evaluate the contribution of reactive oxygen intermediate formation for receptor modulation on neutrophils by the cellulosic dialyzer membrane cuprophan (CU). In patients dialyzed with CU, CD11b and CD66b upregulation on neutrophils (by 104.3 +/- 37.9% and 85.7 +/- 31.1%, respectively), and a downregulation of L-selectin (by 44.9 +/- 26.9%) was seen, whereas expression of CD11a remained unaltered. Hemodialysis with polysulfone did not bring about major changes in surface receptor expression. In vitro incubation of isolated neutrophils in the presence of serum with hollow fibers of CU or polysulfone showed similar results: Only CU resulted in upregulation of CD11b and CD66b expression (by 65.5 +/- 18.7% and 60.1 +/- 24%) and a decrease in CD62L expression (by 60.6 +/- 18.2%). In contrast to receptor alterations, generation of reactive oxygen intermediate by CU occurred in the absence of serum. Inhibition experiments with soluble complement receptor 1, which produced only partial inhibition of receptor up-/down-regulation, indicated the existence of also other than alternate complement-dependent mechanisms for neutrophil activation. By using C5-depleted serum instead of normal human serum, up-/down-regulation of CD11b, CD62L, and CD66b by CU was dramatically reduced, whereas C3-depleted serum did not produce that effect. C5-deficient serum repleted with purified C5, as well as purified C5 alone, was able to induce receptor modulation by CU comparable to normal human serum. L-Methionine, a specific inhibitor for the oxidative activation of C5, blocked the modulatory effect of CU in assays with purified C5 as well as with serum. As a result, in addition to the alternative pathway of complement, a C5-dependent mechanism probably activated by neutrophil-derived reactive oxygen intermediate leads to receptor modulation and subsequent generation of the well known side effects of bioincompatible dialyzer membranes.


Subject(s)
Cellulose/analogs & derivatives , Complement C5/pharmacology , Membranes, Artificial , Neutrophil Activation , Neutrophils/drug effects , Receptors, Cell Surface/analysis , Renal Dialysis , Adult , Aged , Cell Separation , Cells, Cultured , Complement Activation , Complement C5/chemistry , Female , Humans , Male , Middle Aged , Neutrophils/metabolism , Neutrophils/physiology , Polymers , Receptors, Cell Surface/immunology , Receptors, Complement/immunology , Respiratory Burst , Sulfones
11.
Ocul Immunol Inflamm ; 5(1): 11-7, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9145688

ABSTRACT

Leukocytes from ten allergic patients (five allergic to dust-mites and five allergic to pollen) were treated with N-acetyl aspartyl glutamic acid (NAAGA) 4.9%, disodium cromoglycate (DSCG) 2%, lodoxamide (LODO) 1%, and levocabastine (LEVO) 0.5% (concentrations representing the pharmaceutical eyedrop preparations) for 20 minutes. Degranulation was then induced with Complement (rHu5Ca). Histamine was measured in the supernatant with ELISA. LODO and LEVO were inactive in blocking histamine released from human cells, and paradoxical unexpected effects were found with these two agents. They both induced significant histamine release in almost 100% of the samples. DSCG was able to block histamine release in seven patients out of nine (ranging between 5 and 34%). NAAGA was the most active agent on human cells and was able to block basophil degranulation in nine patients out of nine (inhibition ranging between 4 and 66% of total histamine pool).


Subject(s)
Anti-Allergic Agents/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Basophils/physiology , Cell Degranulation/physiology , Complement C5/pharmacology , Histamine H1 Antagonists/pharmacology , Histamine Release , Allergens/adverse effects , Animals , Basophil Degranulation Test , Basophils/drug effects , Cromolyn Sodium/pharmacology , Dipeptides/pharmacology , Enzyme-Linked Immunosorbent Assay , Histamine/analysis , Histamine Release/drug effects , Humans , Hypersensitivity, Immediate/etiology , Hypersensitivity, Immediate/immunology , Ophthalmic Solutions , Oxamic Acid/analogs & derivatives , Oxamic Acid/pharmacology , Piperidines/pharmacology , Recombinant Proteins
12.
Transfusion ; 37(2): 150-9, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9051089

ABSTRACT

BACKGROUND: Liposome-encapsulated hemoglobin (LEH) has been developed as an emergency blood substitute, yet its effect on human complement has never been explored. Considering that complement activation is a major pathogenic factor in the respiratory distress syndrome that often develops in trauma and shock, LEH-induced complement activation may be a critical safety issue. STUDY DESIGN AND METHODS: Various LEH and corresponding empty liposomes were incubated with normal human sera, and various markers of complement activation (serum levels of C4d, Bb, SC5b-9, and CH50; C5a-induced granulocyte aggregation; membrane deposition of C3b) were measured. Incubations were also performed in the presence of (ethylene-bis[oxyethylenenitrilo]tetraacetic acid) (EGTA) and Mg++ (EGTA/Mg++) and soluble complement receptor type 1. RESULTS: LEH and liposomes activated human complement, as indicated by significant changes in one or more markers. The effect was primarily due to the presence of the phospholipid vehicle; small, unilamellar, highly homodispersed vesicles induced the greatest degree of complement activation. Complement activation was partially inhibited by EGTA/Mg++. The latter finding, together with the parallel increases in serum C4d and Bb, suggests activation of both the classical and alternative pathways. Soluble complement receptor type 1 (0.05-20 micrograms/mL) efficiently inhibited all vesicle-induced complement activation. CONCLUSION: Because of complement activation, the use of LEH for transfusion may require careful evaluation of safety. Soluble complement receptor type 1 may be useful as a prophylactic agent for complement activation-related complications of liposome infusions.


Subject(s)
Blood Substitutes/pharmacology , Complement Activation/drug effects , Hemoglobins/administration & dosage , Receptors, Complement/antagonists & inhibitors , Complement C4/pharmacology , Complement C5/pharmacology , Complement C6/pharmacology , Dose-Response Relationship, Drug , Drug Carriers , Egtazic Acid/pharmacology , Humans , Liposomes , Magnesium/pharmacology , Recombinant Proteins/antagonists & inhibitors , Solubility
13.
Int Arch Allergy Immunol ; 110(4): 314-7, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8768797

ABSTRACT

S-protein/vitronectin participates in the regulation of complement-mediated lysis by incorporating into the membrane attack complex C5b-9. Subsequently, soluble SC5b-7 and SC5b-9 macromolecules are not inserted into the membrane lipid bilayer nor induce lytic pore formation. Using a dot blot binding assay, it was determined that soluble S-protein/vitronectin interacted with immobilized C5b and C8 among the five separately immobilized components (C5b, C6, C7, C8, C9) of the membrane attack complex. Those interactions imply a new model of the formation of the complement membrane attack complex and its regulation by S-protein/vitronectin.


Subject(s)
Complement C5/metabolism , Complement C8/metabolism , Complement Membrane Attack Complex/metabolism , Vitronectin/metabolism , Animals , Complement C5/pharmacology , Complement C5b , Complement C8/pharmacology , Heparin/pharmacology , Solubility , Vitronectin/antagonists & inhibitors
14.
Exp Nephrol ; 2(5): 299-305, 1994.
Article in English | MEDLINE | ID: mdl-7812736

ABSTRACT

The complement-regulatory factor C8 binding protein (C8bp) was first identified on human erythrocyte membranes by its affinity for the complement component C8 and its ability to inhibit lysis by homologous complement. Cultured human glomerular mesangial or epithelial cells (GEC) expressed C8bp on the cell surface and in the cytoplasm. Following stimulation of the glomerular cells with interleukin 1 beta, C5b-9 or with endotoxin, a transient, protein synthesis-independent increase in C8bp surface expression was seen. Blocking of C8bp function with F(ab)2 fragment of an antibody to C8bp rendered GEC susceptible to complement-mediated killing, indicating that C8bp contributes to the cellular defense against complement attack.


Subject(s)
Blood Proteins/analysis , CD59 Antigens , Carrier Proteins/analysis , Complement System Proteins/pharmacology , Glomerular Mesangium/chemistry , Glomerular Mesangium/cytology , Blood Proteins/physiology , Carrier Proteins/physiology , Cell Death/drug effects , Cells, Cultured , Complement Activation/physiology , Complement C5/pharmacology , Complement C5b , Endotoxins/pharmacology , Epithelial Cells , Epithelium/drug effects , Glomerular Mesangium/drug effects , Humans , Interleukin-1/pharmacology , Kidney Glomerulus/chemistry , Kidney Glomerulus/cytology
15.
J Leukoc Biol ; 47(5): 449-56, 1990 May.
Article in English | MEDLINE | ID: mdl-2335754

ABSTRACT

The influence of the three PAF-antagonists WEB-2086, L-652,731, and SRI-63441 on the chemotactic response of neutrophil and eosinophil granulocytes to PAF was investigated. When the PAF-antagonists were added to the cell suspension that was exposed to a gradient of PAF, WEB-2086 and SRI-63441 at the concentration of 10(-6) mol/litre inhibited (P less than .01) the neutrophil and eosinophil chemotactic response to 10(-8) and 10(-9) mol PAF per litre; at the concentration of 5 x 10(-6) mol/litre, WEB-2086 and SRI-63441 also inhibited (P less than .02) the response to 10(-7) mol PAF per litre. Under the same conditions L-652,731 at the concentration of 5 x 10(-6) mol/litre inhibited (P less than .01) the eosinophil chemotactic response to 10(-8) and 10(-9) mol PAF per litre. The inhibition of the chemotactic response to PAF by the three PAF-antagonists was specific, since the chemotactic response to C5f, f-MLP, and LTB4 was not affected by WEB-2086, L-652,731, or SRI-63441, neither was the chemokinetic migration induced by albumin.


Subject(s)
Azepines/pharmacology , Chemotaxis/drug effects , Eosinophils/physiology , Furans/pharmacology , Neutrophils/physiology , Platelet Activating Factor/pharmacology , Quinolinium Compounds/pharmacology , Triazoles/pharmacology , Chemotaxis/physiology , Complement C5/pharmacology , Eosinophils/drug effects , Humans , Leukotrienes/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Platelet Activating Factor/antagonists & inhibitors
16.
J Invest Dermatol ; 93(2): 195-200, 1989 Aug.
Article in English | MEDLINE | ID: mdl-2754274

ABSTRACT

We previously reported that complement-binding antibasement membrane zone (BMZ) autoantibodies can mediate complement-dependent directed migration and adherence of leukocytes to the BMZ in cryostat skin sections and that there is heterogeneity in the ability of anti-BMZ autoantibodies to mediate that response. Those observations suggested that directed migration and adherence of leukocytes to the BMZ might be dependent on the amount of complement-activating autoantibody deposited at the BMZ and the extent to which those antibodies could activate complement and generate C5-derived peptides (C5a, C5a des arg). In this study, we have examined the role of autoantibody concentration and C5 in mediating the adherence response. When cryostat skin sections were pretreated with anti-BMZ autoantibodies and subsequently incubated with neutrophils suspended in fresh serum, neutrophils adhered to the BMZ. Adherence was anti-BMZ autoantibody specific and proportional to anti-BMZ autoantibody concentration. To determine the role of C5 in mediating adherence, neutrophils were suspended in increasing concentrations of: 1) fresh serum, 2) heat-inactivated serum, 3) serum pretreated with antihuman C5, 4) serum pretreated with antihuman IgG, 5) C5-depleted serum, 6) purified C5, and 7) C5-depleted serum reconstituted with increasing concentrations of purified C5. The suspensions were then incubated with autoantibody-treated skin sections. The results showed a dose-dependent requirement for fresh serum and for C5-depleted serum reconstituted with increasing doses of C5. Adherence could be detected with C5 concentrations less than 200 ng/ml, which correspond to a C5a/C5a des arg concentration of 10(-8)-10(-9) molar. These results suggest that complement-dependent neutrophil adherence is a highly sensitive method for detecting and quantitating the ability of tissue-deposited anti-BMZ autoantibodies to activate complement and generate C5-derived bioactive peptides, for estimating the amount of C-activating anti-BMZ autoantibody deposited at the BMZ in vivo, and for evaluating the potential role of C-activating anti-BMZ autoantibodies in the pathogenesis of lesions.


Subject(s)
Autoantibodies/immunology , Basement Membrane/immunology , Complement C5/immunology , Skin/immunology , Antibody Specificity , Basement Membrane/physiology , Blood Physiological Phenomena , Cell Adhesion , Complement C5/pharmacology , Humans , Neutrophils/physiology , Osmolar Concentration , Reference Values
17.
J Leukoc Biol ; 45(6): 558-71, 1989 Jun.
Article in English | MEDLINE | ID: mdl-2470847

ABSTRACT

We have examined the release of histamine and LTC4 from purified human basophils challenged with several different stimuli, both physiological and nonphysiological. Basophils (n = 16) challenged with 0.1 micrograms/ml anti-IgE released 38 +/- 4% of their available histamine and 39 +/- 12 ng LTC4/10(6) basophils within 15-30 min. F-Met peptide (n = 8) caused the release of 54 +/- 8% histamine and 42 +/- 25 ng LTC4/10(6) basophils within a period of 2-5 min. C5a caused the release of 22 +/- 3% histamine from selected donors but failed to initiate any LTC4 release unless combined with D2O or 5 mM extracellular calcium. The two nonphysiological stimuli A23187 and TPA caused extensive histamine release, 67 +/- 8 and 82 +/- 11%, respectively, and while A23187 initiated a large and rapid release of leukotriene, TPA failed to release any LTC4 even when combined with D2O or 2-5 mM extracellular calcium. Increased concentrations of extracellular calcium enhanced anti-IgE and f-Met peptide induced release of LTC4 but inhibited the A23187 induced release of leukotriene. A single peak of immunoreactive leukotriene C4 that comigrated with the authentic standard was identified using HPLC followed by radioimmunoassay. No LTD4 or LTE4 could be detected. Purified human basophils incubated with 0.2 microM [3H]AA incorporated 290 pmol/10(6) cells, or 32 +/- 5% of the available label within 60 min. The [3H]AA was taken principally into the phospholipids (73 +/- 5%), with 20 +/- 3% as neutral lipid, and only 5 +/- 2% remaining as the free acid. Three phospholipid subclasses, phosphatidylcholine, PC (24 +/- 2%), phosphatidylinositol, PI (22 +/- 1%), and phosphatidylethanolamine, PE (15 +/- 3%), accounted for the majority of the incorporated [3H]AA while the remainder of the phospholipids accounted for less than 5% of the total cpm. HPLC analysis of the lipid mediators released during stimulation with 0.1 micrograms/ml anti-IgE revealed [3H]LTC4 (2.4 +/- 1.0%), [3H]5HETE (1.0 +/- 0.1%), unmetabolized [3H]AA (91 +/- 2%), and an unidentified peak (3.4 +/- 1.4%). The unknown metabolite eluted with the prostaglandins, was inhibited by indomethacin, and appeared to have a relatively high specific activity. It may thus represent an artifact of the labeling procedure rather than a novel basophil-derived prostaglandin.


Subject(s)
Arachidonic Acids/metabolism , Basophils/metabolism , Calcium/pharmacology , Complement C5/pharmacology , Complement C5a , Deuterium/pharmacology , Histamine Release/drug effects , Humans , Immunoglobulin E/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , SRS-A/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
18.
Scand J Immunol ; 29(6): 709-16, 1989 Jun.
Article in English | MEDLINE | ID: mdl-2544988

ABSTRACT

Functions of eosinophils and neutrophils isolated from normal human blood were determined by measuring chemotactic migration and release of beta-glucuronidase. Four well-characterized chemotaxins, the complement fragment C5a, formyl-methionyl-leucyl-phenylalanine (FMLP), platelet-activating factor (PAF), and leukotriene B4 (LTB4) were used as stimuli. Neutrophils showed remarkable chemotactic responses to all four chemotaxins. In contrast, eosinophils showed a significant chemotactic response to C5a and PAF, but only weak responses to FMLP and LTB4. Using these chemotaxins we found the following order of chemotactic potency (maximal number of migrated cells): C5a = LTB4 greater than FMLP greater than PAF for neutrophils and PAF = C5a greater than LTB4 = FMLP for eosinophils. Neutrophils elicited a significant beta-glucuronidase release when stimulated by C5a and FMLP, whereas only small amounts were released with PAF and LTB4. On the other hand, an amount of beta-glucuronidase released from eosinophils comparable to that from neutrophils was elicited only with C5a. FMLP, LTB4, and PAF caused the release of small percentages of beta-glucuronidase. The important cellular functions of eosinophils and neutrophils, chemotaxis and enzyme release, are thought to be controlled by differential responsiveness to stimuli.


Subject(s)
Chemotactic Factors/pharmacology , Eosinophils/drug effects , Neutrophils/drug effects , Adult , Chemotaxis, Leukocyte/drug effects , Complement C5/pharmacology , Complement C5a , Eosinophils/immunology , Glucuronidase/metabolism , Humans , Leukotriene B4/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/immunology , Platelet Activating Factor/pharmacology
19.
Biochem Biophys Res Commun ; 161(2): 520-4, 1989 Jun 15.
Article in English | MEDLINE | ID: mdl-2472137

ABSTRACT

The neuropeptide substance P (SP), a member of the tachykinin family, has stimulatory effects on various cell types at nanomolar concentrations. SP has also direct effects on polymorphonuclear leukocytes (PMNs). However, unlike other cells, stimulation of PMNs requires extremely high concentrations of the peptide (greater than 10 microM), suggesting that direct PMN activation by SP is not physiologically relevant. By measuring primed stimulation of PMNs, we now demonstrate potent synergistic effects of nanomolar doses of SP on the migratory and cytotoxic functions of human PMNs stimulated by fMLP and C5a. This synergism between SP and chemotactic peptides reveals a new regulatory activity of SP and suggests that neurogenic stimuli may prepare neutrophils for an exaggerated inflammatory response to other phlogistic mediators.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Inflammation/physiopathology , Neutrophils/drug effects , Substance P/pharmacology , Cell Movement/drug effects , Complement C5/pharmacology , Complement C5a , Drug Synergism , Humans , In Vitro Techniques , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Oxygen Consumption/drug effects
20.
J Immunol ; 142(9): 3213-8, 1989 May 01.
Article in English | MEDLINE | ID: mdl-2540238

ABSTRACT

Besides its function as a growth factor, the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) "primes" polymorphonuclear leukocytes (PMN) for enhanced biologic responses to a number of secondary stimuli. We examined the effect of priming PMN with GM-CSF on the production of [3H] platelet-activating factor (PAF) from [3H]acetate upon stimulation with the chemotactic factors FMLP and C5a. In PMN stimulated with the individual peptide mediators alone [3H]PAF levels were close to controls, whereas considerable amounts of [3H]PAF are formed after stimulation of PMN which have been preexposed to GM-CSF. The priming effect was concentration and time dependent. It was optimal after a preincubation period of 2 h. A maximum of [3H]PAF accumulation is reached within 2.5 min (C5a) and 5.0 min (FMLP) after activation of GM-CSF-primed PMN. In addition, we show that PAF isolated from PMN preincubated with GM-CSF and triggered with chemotactic factors is able to enhance the respiratory burst in PMN. PAF formed by sequentially activated PMN could contribute to the enhanced oxygen radical production and cytotoxicity in effector cells and play a role in modulating and amplifying inflammatory reactions.


Subject(s)
Colony-Stimulating Factors/pharmacology , Complement C5/pharmacology , Growth Substances/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/metabolism , Platelet Activating Factor/biosynthesis , Adult , Complement C5a , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Neutrophils/drug effects , Platelet Activating Factor/pharmacology , Platelet Activating Factor/physiology , Recombinant Proteins/pharmacology , Superoxides/biosynthesis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...