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
2.
Front Immunol ; 10: 1639, 2019.
Article in English | MEDLINE | ID: mdl-31379839

ABSTRACT

Background: Eculizumab blocks the lytic complement pathway by inhibiting C5 and has become the standard of care for certain complement-mediated diseases. Previously, we have shown that strong complement activation in vitro overrides the C5 inhibition by Eculizumab, which accounts for residual terminal pathway activity. Results: Here we show that the levels of residual hemolysis in ex vivo assays differ markedly (up to 3.4-fold) across sera collected from different paroxysmal nocturnal hemoglobinuria (PNH) patients on Eculizumab treatment. This large variability of residual activity was also found in sera of healthy donors, thus cross-validating the findings in patients. While PNH patients with residual lytic activities of 11-30% exhibited hemolysis levels around the upper limit of normal (i.e., plasma LDH of ~250 u/L), as expected for PNH patients on Eculizumab therapy, we found sustained and markedly increased LDH levels of around 400 u/L for the patient with the highest residual activity of 37%. Furthermore, the clinical history of nine out of 14 PNH patients showed intravascular breakthrough hemolysis at the time of documented infections despite ample amounts of administered Eculizumab and/or experimentally determined excess over C5. Conclusion: The occurrence of extraordinary high levels of residual terminal pathway activity in PNH patients receiving Eculizumab is rare, but can impair the suppression of hemolysis. The commonly observed low levels of residual terminal pathway activity seen for most PNH patients can exacerbate during severe infections and, thus, can cause pharmacodynamic breakthrough hemolysis in PNH patients treated with Eculizumab.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Complement Activation/drug effects , Complement Inactivating Agents/therapeutic use , Hemoglobinuria, Paroxysmal/blood , Hemoglobinuria, Paroxysmal/drug therapy , Humans
3.
J Immunol ; 202(7): 2082-2094, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30745459

ABSTRACT

The plasma proteins Factor H (FH) and its alternate splice variant FH-like protein 1 (FHL-1) are the major regulators of the complement alternative pathway. The indiscriminate nature of alternative pathway activation necessitates the regulators to be host selective, but the underlying principles of selectivity remained largely elusive. By analyzing human FH and FHL-1 for protection of different host and foreign cells (rabbit and yeast), we uncovered a 2-fold discriminatory mechanism of FH in favor of self: relative to FHL-1, FH exhibits a regulatory benefit on self but importantly, also, a regulatory penalty on nonself surfaces, yielding a selectivity factor of ∼2.4 for sialylated host surfaces. We further show that FHL-1 possesses higher regulatory activity than known but is relatively unselective. The reason for this unexpected high activity of FHL-1 is the observation that the complement regulatory site in FH exceeds the established first four domains. Affinity for C3b, cofactor and decay-accelerating activities, and serum assays demonstrate that the regulatory site extends domains 1-4 and includes domains 5-7. But unlike FH, FHL-1 exhibits a fast plasma clearance in mice, occurs sparsely in human plasma (at one fortieth of the FH concentration), and resists deregulation by FH-related proteins. These physiological differences and its late phylogenetic occurrence argue that FHL-1 is crucial for local rather than systemic compartments. In conclusion, we demonstrate a 2-fold discriminatory power of FH to promote selectivity for self over foreign and show that FHL-1 is more active than known but specialized for regulation on local tissues.


Subject(s)
Complement Pathway, Alternative/immunology , Self Tolerance/immunology , Animals , Complement Activation/immunology , Complement Factor H/immunology , Humans
4.
Transfusion ; 58(12): 2992-3002, 2018 12.
Article in English | MEDLINE | ID: mdl-30367826

ABSTRACT

BACKGROUND: Therapeutic intervention strategies in complement-mediated hemolytic diseases are still inappropriate, and lethal events cannot be reliably prevented. As an in vitro model of intravascular hemolysis, a sensitive flow cytometric assay was designed using red blood cells (RBCs) of patients with paroxysmal nocturnal hemoglobinuria (PNH) as target cells. Complement activation by human allo- and autoantibodies directed against RBC antigens and the effect of different complement inhibitors were studied. STUDY DESIGN AND METHODS: RBCs of patients with a PNH III RBC clone of more than 20% were coated with different human allo- or autoantibodies. Hemolysis was initiated with pooled normal human AB serum with or without the addition of complement inhibitors. Loss of PNH III RBCs was estimated by flow cytometry. RESULTS: RBC antibodies of 174 different patients representing 37 different specificities were tested for their potency to activate complement. In correlation with blood group specificities roughly three different patterns were observed: 1) strong and regular, 2) sporadic, and 3) weak or absent complement activation. Remarkably strong complement activators were among antibodies directed against high-prevalence blood group antigens. The C5 inhibitor eculizumab abrogated mild but not strong complement activation, even in presence of excess inhibitor. However, this residual complement activity could be further depressed by combining eculizumab with other inhibitors. CONCLUSION: The PNH hemolysis assay offers a sensitive tool for in vitro analyses of classical pathway-mediated complement activation. The recognition of additive effects of complement inhibitors may guide novel intervention strategies against unwanted complement damage.


Subject(s)
Autoantibodies/immunology , Complement Activation/immunology , Complement Inactivating Agents/pharmacology , Erythrocytes/immunology , Flow Cytometry , Hemoglobinuria, Paroxysmal/immunology , Hemolysis/immunology , Isoantibodies/immunology , Complement Activation/drug effects , Erythrocytes/pathology , Female , Hemoglobinuria, Paroxysmal/pathology , Hemolysis/drug effects , Humans , Male
5.
Glycobiology ; 28(10): 765-773, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29982679

ABSTRACT

Complement factor H (FH), an elongated and substantially glycosylated 20-domain protein, is a soluble regulator of the complement alternative pathway (AP). It contains several glycan binding sites which mediate recognition of α2-3-linked sialic acid (FH domain 20) and glycosaminoglycans (domains 6-8 and 19-20). FH also binds the complement C3-activation product C3b, a powerful opsonin and focal point for the formation of C3-convertases of the AP feedback loop. In freely circulating FH the C3b binding site in domains 19-20 is occluded, a phenomenon that is not fully understood and could be mediated by an intramolecular interaction between FH's intrinsic sialylated glycosylation and its own sialic acid binding site. In order to assess this possibility, we characterized FH's sialylation with respect to glycosidic linkage type and searched for further potential, not yet characterized sialic acid binding sites in FH and its seven-domain spanning splice variant and fellow complement regulator FH like-1 (FHL-1). We also probed FH binding to the sialic acid variant Neu5Gc which is not expressed in humans but on heterologous erythrocytes that restrict the human AP and in FH transgenic mice. We find that FH contains mostly α2-6-linked sialic acid, making an intramolecular interaction with its α2-3-sialic acid specific binding site and an associated self-lock mechanism unlikely, substantiate that there is only a single sialic acid binding site in FH and none in FHL-1, and demonstrate direct binding of FH to the nonhuman sialic acid Neu5Gc, supporting the use of FH transgenic mouse models for studies of complement-related diseases.


Subject(s)
N-Acetylneuraminic Acid/analysis , Animals , Binding Sites , Carbohydrate Conformation , Complement Factor H/chemistry , Complement Factor H/isolation & purification , Complement Factor H/metabolism , Humans , Mice , Mice, Transgenic , N-Acetylneuraminic Acid/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism
6.
Blood ; 129(8): 970-980, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28028023

ABSTRACT

Eculizumab inhibits the terminal, lytic pathway of complement by blocking the activation of the complement protein C5 and shows remarkable clinical benefits in certain complement-mediated diseases. However, several reports suggest that activation of C5 is not always completely suppressed in patients even under excess of eculizumab over C5, indicating that residual C5 activity may derogate the drug's therapeutic benefit under certain conditions. By using eculizumab and the tick-derived C5 inhibitor coversin, we determined conditions ex vivo in which C5 inhibition is incomplete. The degree of such residual lytic activity depended on the strength of the complement activator and the resulting surface density of the complement activation product C3b, which autoamplifies via the alternative pathway (AP) amplification loop. We show that at high C3b densities required for binding and activation of C5, both inhibitors reduce but do not abolish this interaction. The decrease of C5 binding to C3b clusters in the presence of C5 inhibitors correlated with the levels of residual hemolysis. However, by employing different C5 inhibitors simultaneously, residual hemolytic activity could be abolished. The importance of AP-produced C3b clusters for C5 activation in the presence of eculizumab was corroborated by the finding that residual hemolysis after forceful activation of the classical pathway could be reduced by blocking the AP. By providing insights into C5 activation and inhibition, our study delivers the rationale for the clinically observed phenomenon of residual terminal pathway activity under eculizumab treatment with important implications for anti-C5 therapy in general.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Complement Activation/drug effects , Complement C5/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/therapeutic use , Complement C3b/immunology , Complement C5/immunology , Complement Pathway, Alternative/drug effects , Hemoglobinuria, Paroxysmal/drug therapy , Hemoglobinuria, Paroxysmal/immunology , Hemolysis/drug effects , Humans
7.
Immunobiology ; 221(4): 503-11, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26792457

ABSTRACT

Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by complement-mediated cell lysis due to deficiency of GPI-anchored complement regulators. Blockage of the lytic pathway by eculizumab is the only available therapy for PNH patients and shows remarkable benefits, but regularly yields PNH erythrocytes opsonized with fragments of complement protein C3, rendering such erythrocytes prone to extravascular hemolysis. This effect is associated with insufficient responsiveness seen in a subgroup of PNH patients. Novel C3-opsonin targeted complement inhibitors act earlier in the cascade, at the level of activated C3 and are engineered from parts of the natural complement regulator Factor H (FH) or complement receptor 2 (CR2). This inhibitor class comprises three variants of "miniFH" and the clinically developed "FH-CR2" fusion-protein (TT30). We show that the approach of FH-CR2 to target C3-opsonins was more efficient in preventing complement activation induced by foreign surfaces, whereas the miniFH variants were substantially more active in controlling complement on PNH erythrocytes. Subtle differences were noted in the ability of each version of miniFH to protect human PNH cells. Importantly, miniFH and FH-CR2 interfered only minimally with complement-mediated serum killing of bacteria when compared to untargeted inhibition of all complement pathways by eculizumab. Thus, the molecular design of each C3-opsonin targeted complement inhibitor determines its potency in respect to the nature of the activator/surface providing potential functionality in PNH.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Complement C3/genetics , Complement Inactivating Agents/pharmacology , Erythrocytes/drug effects , Opsonin Proteins/genetics , Animals , Antibodies, Monoclonal, Humanized/biosynthesis , Antibodies, Monoclonal, Humanized/immunology , Cells, Cultured , Complement Activation/drug effects , Complement Factor H/genetics , Complement Factor H/immunology , Complement Inactivating Agents/immunology , Complement Inactivating Agents/metabolism , Complement Pathway, Alternative , Erythrocytes/immunology , Erythrocytes/pathology , Escherichia coli/drug effects , Escherichia coli/growth & development , Gene Expression , Hemoglobinuria, Paroxysmal/drug therapy , Hemoglobinuria, Paroxysmal/genetics , Hemoglobinuria, Paroxysmal/metabolism , Hemoglobinuria, Paroxysmal/pathology , Hemolysis/drug effects , Humans , Molecular Targeted Therapy , Protein Engineering , Rabbits , Receptors, Complement 3d/genetics , Receptors, Complement 3d/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
8.
J Immunol ; 196(2): 866-76, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26643478

ABSTRACT

The serum proteins factor H (FH), consisting of 20 complement control protein modules (CCPs), and its splice product FH-like protein 1 (FHL-1; consisting of CCPs 1-7) are major regulators of the alternative pathway (AP) of complement activation. The engineered version of FH, miniFH, contains only the N- and C-terminal portions of FH linked by an optimized peptide and shows ∼ 10-fold higher ex vivo potency. We explored the hypothesis that regulatory potency is enhanced by unmasking of a ligand-binding site in the C-terminal CCPs 19-20 that is cryptic in full-length native FH. Therefore, we produced an FH variant lacking the central domains 10-15 (FHΔ10-15). To explore how avidity affects regulatory strength, we generated a duplicated version of miniFH, termed midiFH. We compared activities of FHΔ10-15 and midiFH to miniFH, FH, and FHL-1. Relative to FH, FHΔ10-15 exhibited an altered binding profile toward C3 activation products and a 5-fold-enhanced complement regulation on a paroxysmal nocturnal hemoglobinuria patient's erythrocytes. Contrary to dogma, FHL-1 and FH exhibited equal regulatory activity, suggesting that the role of FHL-1 in AP regulation has been underestimated. Unexpectedly, a substantially increased avidity for complement opsonins, as seen in midiFH, did not potentiate the inhibitory potential on host cells. In conclusion, comparisons of engineered and native FH-based regulators have identified features that determine high AP regulatory activity on host cells. Unrestricted availability of FH CCPs 19-20 and an optimal spatial orientation between the N- and C-terminal FH regions are key.


Subject(s)
Complement C3b Inactivator Proteins/immunology , Complement Factor H/immunology , Complement Inactivating Agents/pharmacology , Complement Pathway, Alternative/immunology , Recombinant Proteins/pharmacology , Amino Acid Sequence , Complement Factor H/chemistry , Complement Inactivating Agents/chemical synthesis , Complement Inactivating Agents/immunology , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Protein Binding , Recombinant Proteins/chemical synthesis , Recombinant Proteins/immunology
9.
J Biol Chem ; 290(42): 25307-21, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26324715

ABSTRACT

Plasmodium falciparum parasites must invade red blood cells to survive within humans. Entry into red blood cells is governed by interactions between parasite adhesins and red blood cell receptors. Previously we identified that P. falciparum reticulocyte binding protein-like homologue 4 (PfRh4) binds to complement receptor 1 (CR1) to mediate entry of malaria parasites into human red blood cells. In this report we characterize a collection of anti-PfRh4 monoclonal antibodies and CR1 protein fragments that modulate the interaction between PfRh4 and CR1. We identify an anti-PfRh4 monoclonal that blocks PfRh4-CR1 interaction in vitro, inhibits PfRh4 binding to red blood cells, and as a result abolishes the PfRh4-CR1 invasion pathway in P. falciparum. Epitope mapping of anti-PfRh4 monoclonal antibodies identified distinct functional regions within PfRh4 involved in modulating its interaction with CR1. Furthermore, we designed a set of protein fragments based on extensive mutagenesis analyses of the PfRh4 binding site on CR1 and determined their interaction affinities using surface plasmon resonance. These CR1 protein fragments bind tightly to PfRh4 and also function as soluble inhibitors to block PfRh4 binding to red blood cells and to inhibit the PfRh4-CR1 invasion pathway. Our findings can aid future efforts in designing specific single epitope antibodies to block P. falciparum invasion via complement receptor 1.


Subject(s)
Antibodies, Monoclonal/immunology , Erythrocytes/metabolism , Membrane Proteins/metabolism , Protozoan Proteins/metabolism , Receptors, Complement/metabolism , Animals , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...