Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 314
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Immunol ; 213(2): 235-243, 2024 07 15.
Article in English | MEDLINE | ID: mdl-38819221

ABSTRACT

The development of agonists capable of activating the human complement system by binding to the C1 complex presents a novel approach for targeted cell killing. Bispecific nanobodies and Abs can successfully use C1 for this purpose; however, efficacy varies significantly between epitopes, Ab type, and bispecific design. To address this variability, we investigated monomeric agonists of C1 in the form of bispecific nanobodies, which lack Fc domains that lead to oligomerization in Abs. These therefore offer an ideal opportunity to explore the geometric parameters crucial for C1 activation. In this study, we explored the impact of linker length as a metric for Ag and epitope location. DNA nanotechnology and protein engineering allowed us to design linkers with controlled lengths and flexibilities, revealing a critical range of end-to-end distances for optimal complement activation. We discovered that differences in complement activation were not caused by differential C1 activation or subsequent cleavage of C4, but instead impacted C4b deposition and downstream membrane lysis. Considering the importance of Ab class and subclass, this study provides insights into the structural requirements of C1 binding and activation, highlighting linker and hinge engineering as a potential strategy to enhance potency over specific cellular targets. Additionally, using DNA nanotechnology to modify geometric parameters demonstrated the potential for synthetic biology in complement activation. Overall, this research offers valuable insights into the design and optimization of agonists for targeted cell killing through complement activation.


Subject(s)
Antibodies, Bispecific , Complement Activation , Protein Engineering , Humans , Complement Activation/immunology , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Complement C1/immunology , Single-Domain Antibodies/immunology , Epitopes/immunology , Protein Binding , Complement C4b/immunology
2.
Proc Natl Acad Sci U S A ; 116(24): 11900-11905, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31147461

ABSTRACT

Antigen binding by serum Ig-M (IgM) protects against microbial infections and helps to prevent autoimmunity, but causes life-threatening diseases when mistargeted. How antigen-bound IgM activates complement-immune responses remains unclear. We present cryoelectron tomography structures of IgM, C1, and C4b complexes formed on antigen-bearing lipid membranes by normal human serum at 4 °C. The IgM-C1-C4b complexes revealed C4b product release as the temperature-limiting step in complement activation. Both IgM hexamers and pentamers adopted hexagonal, dome-shaped structures with Fab pairs, dimerized by hinge domains, bound to surface antigens that support a platform of Fc regions. C1 binds IgM through widely spread C1q-collagen helices, with C1r proteases pointing outward and C1s bending downward and interacting with surface-attached C4b, which further interacts with the adjacent IgM-Fab2 and globular C1q-recognition unit. Based on these data, we present mechanistic models for antibody-mediated, C1q-transmitted activation of C1 and for C4b deposition, while further conformational rearrangements are required to form C3 convertases.


Subject(s)
Complement Activation/immunology , Complement C1/immunology , Complement C4/immunology , Immunoglobulin M/immunology , Antibodies/immunology , Antigens/immunology , Binding Sites/immunology , Humans , Models, Molecular
3.
Int J Mol Sci ; 23(4)2022 Feb 14.
Article in English | MEDLINE | ID: mdl-35216207

ABSTRACT

Immunoglobulin G (IgG) adopts a modular multidomain structure that mediates antigen recognition and effector functions, such as complement-dependent cytotoxicity. IgG molecules are self-assembled into a hexameric ring on antigen-containing membranes, recruiting the complement component C1q. In order to provide deeper insights into the initial step of the complement pathway, we report a high-speed atomic force microscopy study for the quantitative visualization of the interaction between mouse IgG and the C1 complex composed of C1q, C1r, and C1s. The results showed that the C1q in the C1 complex is restricted regarding internal motion, and that it has a stronger binding affinity for on-membrane IgG2b assemblages than C1q alone, presumably because of the lower conformational entropy loss upon binding. Furthermore, we visualized a 1:1 stoichiometric interaction between C1/C1q and an IgG2a variant that lacks the entire CH1 domain in the absence of an antigen. In addition to the canonical C1q-binding site on Fc, their interactions are mediated through a secondary site on the CL domain that is cryptic in the presence of the CH1 domain. Our findings offer clues for novel-modality therapeutic antibodies.


Subject(s)
Complement C1/immunology , Immunoglobulin G/immunology , Protein Binding/immunology , Animals , Binding Sites/immunology , Complement Activation/immunology , Mice
4.
Nano Lett ; 19(7): 4787-4796, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31184907

ABSTRACT

IgG antibodies play a central role in protection against pathogens by their ability to alert and activate the innate immune system. Here, we show that IgGs assemble into oligomers on antigenic surfaces through an ordered, Fc domain-mediated process that can be modulated by protein engineering. Using high-speed atomic force microscopy, we unraveled the molecular events of IgG oligomer formation on surfaces. IgG molecules were recruited from solution although assembly of monovalently binding molecules also occurred through lateral diffusion. Monomers were observed to assemble into hexamers with all intermediates detected, but in which only hexamers bound C1. Functional characterization of oligomers on cells also demonstrated that C1 binding to IgG hexamers was a prerequisite for maximal activation, whereas tetramers, trimers, and dimers were mostly inactive. We present a dynamic IgG oligomerization model, which provides a framework for exploiting the macromolecular assembly of IgGs on surfaces for tool, immunotherapy, and vaccine design.


Subject(s)
Complement Activation , Complement C1/chemistry , Immunoglobulin G/chemistry , Protein Multimerization , Complement C1/immunology , Humans , Immunoglobulin G/immunology
5.
PLoS Pathog ; 13(7): e1006531, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28742139

ABSTRACT

Whooping cough, or pertussis, is a contagious disease of the respiratory tract that is re-emerging worldwide despite high vaccination coverage. The causative agent of this disease is the Gram-negative Bordetella pertussis. Knowledge on complement evasion strategies of this pathogen is limited. However, this is of great importance for future vaccine development as it has become apparent that a novel pertussis vaccine is needed. Here, we unravel the effect of Virulence associated gene 8 (Vag8) of B. pertussis on the human complement system at the molecular level. We show that both recombinant and endogenously secreted Vag8 inhibit complement deposition on the bacterial surface at the level of C4b. We reveal that Vag8 binding to human C1-inhibitor (C1-inh) interferes with the binding of C1-inh to C1s, C1r and MASP-2, resulting in the release of active proteases that subsequently cleave C2 and C4 away from the bacterial surface. We demonstrate that the depletion of these complement components in the bacterial surrounding and subsequent decreased deposition on B. pertussis leads to less complement-mediated bacterial killing. Vag8 is the first protein described that specifically prevents C1s, C1r and MASP-2 binding to C1-inh and thereby mediates complement consumption away from the bacterial surface. Unravelling the mechanism of this unique complement evasion strategy of B. pertussis is one of the first steps towards understanding the interactions between the first line of defense complement and B. pertussis.


Subject(s)
Bacterial Proteins/immunology , Bordetella pertussis/immunology , Complement C1/immunology , Complement C2/immunology , Complement C4/immunology , Virulence Factors, Bordetella/immunology , Whooping Cough/immunology , Bacterial Proteins/genetics , Bordetella pertussis/genetics , Humans , Virulence , Virulence Factors, Bordetella/genetics , Whooping Cough/microbiology
6.
PLoS Pathog ; 12(1): e1005404, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26808924

ABSTRACT

Pathogens that traffic in blood, lymphatics, or interstitial fluids must adopt strategies to evade innate immune defenses, notably the complement system. Through recruitment of host regulators of complement to their surface, many pathogens are able to escape complement-mediated attack. The Lyme disease spirochete, Borrelia burgdorferi, produces a number of surface proteins that bind to factor H related molecules, which function as the dominant negative regulator of the alternative pathway of complement. Relatively less is known about how B. burgdorferi evades the classical pathway of complement despite the observation that some sensu lato strains are sensitive to classical pathway activation. Here we report that the borrelial lipoprotein BBK32 potently and specifically inhibits the classical pathway by binding with high affinity to the initiating C1 complex of complement. In addition, B. burgdorferi cells that produce BBK32 on their surface bind to both C1 and C1r and a serum sensitive derivative of B. burgdorferi is protected from killing via the classical pathway in a BBK32-dependent manner. Subsequent biochemical and biophysical approaches localized the anti-complement activity of BBK32 to its globular C-terminal domain. Mechanistic studies reveal that BBK32 acts by entrapping C1 in its zymogen form by binding and inhibiting the C1 subcomponent, C1r, which serves as the initiating serine protease of the classical pathway. To our knowledge this is the first report of a spirochetal protein acting as a direct inhibitor of the classical pathway and is the only example of a biomolecule capable of specifically and noncovalently inhibiting C1/C1r. By identifying a unique mode of complement evasion this study greatly enhances our understanding of how pathogens subvert and potentially manipulate host innate immune systems.


Subject(s)
Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Complement Activation/immunology , Complement Pathway, Classical/immunology , Host-Parasite Interactions/immunology , Immune Evasion/immunology , Complement C1/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Immunoprecipitation , Lyme Disease/immunology
7.
J Immunol ; 188(9): 4450-9, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22467648

ABSTRACT

Ail is a 17-kDa chromosomally encoded outer membrane protein that mediates serum resistance (complement resistance) in the pathogenic Yersiniae (Yersinia pestis, Y. enterocolitica, and Y. pseudotuberculosis). In this article, we demonstrate that Y. pseudotuberculosis Ail from strains PB1, 2812/79, and YPIII/pIB1 (serotypes O:1a, O:1b, and O:3, respectively) can bind the inhibitor of the classical and lectin pathways of complement, C4b-binding protein (C4BP). Binding was observed irrespective of serotype tested and independently of YadA, which is the primary C4BP receptor of Y. enterocolitica. Disruption of the ail gene in Y. pseudotuberculosis resulted in loss of C4BP binding. Cofactor assays revealed that bound C4BP is functional, because bound C4BP in the presence of factor I cleaved C4b. In the absence of YadA, Ail conferred serum resistance to strains PB1 and YPIII, whereas serum resistance was observed in strain 2812/79 in the absence of both YadA and Ail, suggesting additional serum resistance factors. Ail from strain YPIII/pIB1 alone can mediate serum resistance and C4BP binding, because its expression in a serum-sensitive laboratory strain of Escherichia coli conferred both of these phenotypes. Using a panel of C4BP mutants, each deficient in a single complement control protein domain, we observed that complement control protein domains 6-8 are important for binding to Ail. Binding of C4BP was unaffected by increasing heparin or salt concentrations, suggesting primarily nonionic interactions. These results indicate that Y. pseudotuberculosis Ail recruits C4BP in a functional manner, facilitating resistance to attack from complement.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Complement Inactivator Proteins/immunology , Complement Pathway, Classical/immunology , Complement Pathway, Mannose-Binding Lectin/immunology , Histocompatibility Antigens/immunology , Yersinia pseudotuberculosis/immunology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/immunology , Adhesins, Bacterial/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Blood Bactericidal Activity/genetics , Blood Bactericidal Activity/immunology , Complement C1/genetics , Complement C1/immunology , Complement C1/metabolism , Complement C4b-Binding Protein , Complement Inactivator Proteins/genetics , Complement Inactivator Proteins/metabolism , Complement Pathway, Classical/genetics , Complement Pathway, Mannose-Binding Lectin/genetics , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli/metabolism , Female , Histocompatibility Antigens/genetics , Histocompatibility Antigens/metabolism , Humans , Male , Mutation , Protein Binding/genetics , Protein Binding/immunology , Species Specificity , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/metabolism
8.
Int J Immunogenet ; 40(3): 171-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22913810

ABSTRACT

The Luminex xMAP system has become an important tool for HLA antibody screening and identification in sera of transplant patients. Recently, the Luminex single antigen bead assay was shown to be prone to an artefact, the so called prozone phenomenon: Sera with high titer HLA antibodies gave negative results when tested neat, but reacted strongly positive after 1:10 dilution. We also observed such a phenomenon and found that it was most likely caused by the complement component 1 (C1) by competitively displacing the detection antibodies. In this article we review the complement-mediated prozone effect and other mechanisms of interference with solid phase assays, and we discuss possible consequences for HLA antibody testing with the Luminex SAB assay.


Subject(s)
Antibodies/blood , Antibodies/immunology , Complement System Proteins/immunology , HLA Antigens/immunology , Histocompatibility Testing/methods , Histocompatibility Testing/standards , Antigens/immunology , Complement C1/immunology , Complement C4/immunology , Humans , Immunoassay/methods
9.
J Exp Med ; 164(4): 981-97, 1986 Oct 01.
Article in English | MEDLINE | ID: mdl-3760783

ABSTRACT

As part of a broader analysis of the mechanism(s) by which the most sensitive (type III) paroxysmal nocturnal hemoglobinuria (PNH) erythrocytes are excessively sensitive to reactive lysis by isolated C5b6, C7, C8, and C9, we have compared type III PNH (PNH-III) and normal human E in respect to both total specific binding of 125I-C9 and the proportion of cell-bound C9 appearing in high molecular weight (HMW) complexes. In a previous report, we found that after exposure to purified C5b6 and 125I-C7, specific C7 binding and, by implication, EC5b-7 formation were equal for PNH-III E and normal E. In the present study, C8-dependent binding of 125I-C9 to PNH-III EC5b-7 and normal EC5b-7 was also similar, although lysis of the PNH-III E was up to five times greater; that is, PNH-III E required fewer bound C9 molecules to produce an effective lytic site than did normal E. To quantify radioactivity in monomeric and HMW forms of membrane-bound C9, lysed and unlysed E were subjected to low ionic strength buffers to convert all E to ghosts. These ghosts were solubilized in 0.1 or 2% SDS (without reduction) and electrophoresed on 2.4-11% polyacrylamide gradient gels followed by autoradiography and densitometric scanning. With 0.1% SDS, broad, heterodisperse zones of HMW C9 were recovered from both PNH and normal ghosts; the amounts of C9 incorporated into the HMW complexes were similar for PNH-III E and normal E. In selected experiments, 125I-C7 could be shown in these same HMW bands. When membranes were solubilized in 2% SDS, the overall proportion of HMW C9 complexes compared with dimer and monomer C9 was reduced on both PNH and normal membranes. In many, but not all experiments, more of the highest mol wt C9 complexes were detected from PNH-III E membranes solubilized in 2% SDS than from normal or PNH-II E membranes similarly treated. When antibody-sensitized E were lysed by purified C1-C9, PNH-III EA bound far more C9 than did normal EA, and both lysis and C9 incorporation into HMW complexes were markedly and proportionately increased over normal; however, lytic efficiency of 125I-C9 bound to PNH EA was equal to or less than that bound to normal EA.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Complement C9/immunology , Erythrocytes/immunology , Hemoglobinuria, Paroxysmal/immunology , Hemolysis , Antibodies/immunology , Complement C1/immunology , Complement C7/immunology , Complement C8/immunology , Complement Membrane Attack Complex , Complement System Proteins/immunology , Humans , Molecular Weight
10.
J Exp Med ; 166(5): 1351-61, 1987 Nov 01.
Article in English | MEDLINE | ID: mdl-3500259

ABSTRACT

Cell lines have been established that secrete a matched set of human chimeric IgM, IgG1, IgG2, IgG3, IgG4, IgE, and IgA2 antibodies that are directed against the hapten 4-hydroxy-3-nitrophenacetyl. These chimeric antibodies secreted from mouse plasmacytoma cells behave exactly like their authentic human counterparts in SDS-PAGE analysis, binding to protein A and in a wide range of serological assays. The antibodies have been compared in their ability to bind human C1q as well as in their efficacy in mediating lysis of human erythrocytes in the presence of human complement. A major conclusion to emerge is that whereas IgG3 bound C1q better than did IgG1, the chimeric IgG1 was much more effective than all the other IgG subclasses in complement-dependent hemolysis. The IgG1 antibody was also the most effective in mediating antibody-dependent cell-mediated cytotoxicity using both human effector and human target cells. These results suggest that IgG1 might be the favoured IgG subclass for therapeutic applications.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulins/immunology , Animals , Antibodies, Monoclonal/genetics , Antibody-Dependent Cell Cytotoxicity , Cell Line , Complement Activating Enzymes/immunology , Complement C1/immunology , Complement C1q , Complement System Proteins/immunology , DNA, Recombinant , Electrophoresis, Polyacrylamide Gel , Genes, Immunoglobulin , Glycosylation , Haptens , Hemolysis , Humans , Immunoglobulins/genetics , Mice , Nitrophenols/immunology , Phenylacetates , Plasmacytoma/immunology , Plasmids , Transfection , Tumor Cells, Cultured
11.
J Exp Med ; 168(1): 127-42, 1988 Jul 01.
Article in English | MEDLINE | ID: mdl-3260935

ABSTRACT

Humanized antibodies are likely to have a major role in therapy and it is important to define their interaction with physiological effectors. By comparing a matched series of chimeric human mAbs we found that igG1 was most efficient in complement lysis, although IgG3 bound more C1q. To resolve this paradox we compared the ability of human IgG1, IgG2, IgG3, IgG4, and IgE and rat IgG2b to cause C1q binding, C1 binding and activation, C4 activation, C4b binding, and C3b binding. Rat IgG2b was included because this isotype has already successfully been used for therapy. Human IgG1 was less efficient than IgG3 and fixing C1q and C1 on the cell surface, but the number of C4 molecules bound per C1 was 10-fold greater for IgG1 than for IgG3. This difference, amplified through later stages of the complement cascade, can account for the superiority of IgG1 for cell lysis. The efficiency of IgG1 in fixing C4 was not due to a favored binding site on the antibody molecule, since virtually all of the bound C4b was attached to the cells. Rather, it appeared that the activation of C4 by C1s was greatly favored by IgG1 compared with IgG3. It should be possible to combine the optimal properties of IgG1 and IgG3 antibodies to produce an improved therapeutic reagent.


Subject(s)
Antibodies, Monoclonal/immunology , Complement Activating Enzymes/immunology , Complement Activation , Complement C1/immunology , Complement C4/immunology , Immunoglobulin G/immunology , Complement C1q , Complement C3/immunology , Complement C3b/immunology , Complement C4b , Erythrocyte Membrane/immunology , Erythrocytes/immunology , Haptens , Hemolysis , Humans , Immunoglobulin Isotypes/immunology , Nitrohydroxyiodophenylacetate/immunology
12.
BMC Immunol ; 11: 43, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20727163

ABSTRACT

BACKGROUND: Complement C2 deficiency is the most common genetically determined complete complement deficiency and is associated with a number of diseases. Most prominent are the associations with recurrent serious infections in young children and the development of systemic lupus erythematosus (SLE) in adults. The links with these diseases reflect the important role complement C2 plays in both innate immunity and immune tolerance. Infusions with normal fresh frozen plasma for the treatment of associated disease have demonstrated therapeutic effects but so far protein replacement therapy has not been evaluated. RESULTS: Human complement C2 was cloned and expressed in a mammalian cell line. The purity of recombinant human C2 (rhC2) was greater than 95% and it was characterized for stability and activity. It was sensitive to C1s cleavage and restored classical complement pathway activity in C2-deficient serum both in a complement activation ELISA and a hemolytic assay. Furthermore, rhC2 could increase C3 fragment deposition on the human pathogen Streptococcus pneumoniae in C2-deficient serum to levels equal to those with normal serum. CONCLUSIONS: Taken together these data suggest that recombinant human C2 can restore classical complement pathway activity and may serve as a potential therapeutic for recurring bacterial infections or SLE in C2-deficient patients.


Subject(s)
Complement C2/metabolism , Immunologic Deficiency Syndromes/genetics , Lupus Erythematosus, Systemic/genetics , Recombinant Proteins/metabolism , Streptococcal Infections/genetics , Streptococcus pneumoniae/immunology , Adult , Cell Line, Transformed , Child , Complement C1/immunology , Complement C1/metabolism , Complement C2/genetics , Complement C2/therapeutic use , Complement C3/immunology , Complement C3/metabolism , Complement Pathway, Classical/drug effects , Humans , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/drug therapy , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/drug therapy , Protein Binding/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Recurrence , Streptococcal Infections/complications , Streptococcal Infections/drug therapy
13.
Clin Immunol ; 134(3): 354-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19945350

ABSTRACT

The diagnosis of hereditary angioedema (HAE) is based on complement tests. We studied for the first time the possible association between complement parameters measured at the time of diagnosis and disease severity in 115 patients with HAE. Serum levels of functional C1-inhibitor (C1-INH(f)), antigenic C1-inhibitor (C1-INH(a)), C4 and hemolytic activity of the classical pathway (CH50) were determined at the time of diagnosis. We found a significant correlation between severity scores and baseline C1-INH(f) levels, as determined by ELISA assay (p=0.0003). On the other hand, there was no correlation between severity scores and other complement parameters (C1-INH(a), C4, and CH50). We consider the correlation between severity scores and baseline C1-INH(f) levels an important pathophysiological observation. Our findings underlie the potential significance of monitoring functional C1-INH levels in relation to clinical disease course.


Subject(s)
Angioedemas, Hereditary/immunology , Complement C1 Inhibitor Protein/immunology , Adolescent , Adult , Aged , Angioedemas, Hereditary/diagnosis , Child , Child, Preschool , Complement C1/immunology , Complement C1 Inhibitor Protein/analysis , Complement C4/immunology , Complement Hemolytic Activity Assay , Female , Humans , Infant , Male , Middle Aged , Retrospective Studies , Severity of Illness Index , Statistics, Nonparametric , Young Adult
14.
Mol Immunol ; 46(4): 688-94, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18950866

ABSTRACT

BACKGROUND: Meconium aspiration syndrome has a complex pathophysiology. Meconium activates the complement system and meconium-induced cytokine formation is differentially mediated by complement and CD14. C1-inhibitor (C1-INH) regulates complement and contact-system activation mainly by protease inhibition, but may reduce inflammation by other mechanisms as well. OBJECTIVE: The aim of the study was to investigate the initial mechanisms of meconium-induced complement activation and to study the effect of C1-INH on the meconium-induced inflammatory reaction. METHODS: Human serum from five donors was preincubated with an anti-MBL monoclonal antibody and then incubated with meconium for 30 min at 37 degrees C. Human cord whole blood, anticoagulated with lepirudin, from six donors was preincubated with C1-INH and then incubated with meconium for 30 min and 4h at 37 degrees C. Complement activation products specific for the different pathways were measured by ELISAs: classical pathway C1rs/C1-INH complexes, classical and lectin pathway C4d, alternative pathway C3bBbP, and terminal pathway sC5b-9 complex (TCC). A Bio-Plex Array Reader was used to measure 27 inflammatory mediators. RESULTS: The anti-MBL monoclonal antibody significantly reduced meconium-induced formation of C4d by 63% (p=0.0159) and TCC by 27% (p=0.0079). C1-INH dose-dependently inhibited meconium-induced formation of C1rs/C1-INH complexes, C4d, C3bBbP, and TCC compared to albumin (p<0.002 for all). C1-INH induced a dose-dependent and substantial inhibition of meconium-induced formation of the proinflammatory cytokines TNFalpha, IL-1 beta, IL-6 and IFN-gamma (p<0.01 for all), the chemokines IL-8, MCP-1, MIP-1 alpha, MIP-1 beta, and eotaxin (p<0.02 for all), the growth factors G-CSF, GM-CSF, basic FGF, and PDGFbb (p<0.05 for all), and the anti-inflammatory cytokine IL-1ra (p<0.001). CONCLUSIONS: Meconium activated the lectin complement pathway as well as the alternative pathway. C1-INH efficiently reduced a broad spectrum of inflammatory mediators even at the lowest concentration. Administration of C1-INH may thus reduce the inflammatory response in MAS.


Subject(s)
Complement Activation/immunology , Complement C1 Inhibitor Protein/immunology , Complement C1/metabolism , Fetal Blood/immunology , Inflammation/immunology , Meconium/immunology , Complement C1/antagonists & inhibitors , Complement C1/immunology , Complement C1 Inhibitor Protein/pharmacology , Complement Pathway, Alternative/immunology , Cytokines/immunology , Cytokines/metabolism , Fetal Blood/metabolism , Humans , Infant, Newborn , Inflammation/metabolism , Meconium Aspiration Syndrome/immunology , Meconium Aspiration Syndrome/prevention & control
15.
Front Immunol ; 11: 1561, 2020.
Article in English | MEDLINE | ID: mdl-32793217

ABSTRACT

Because of its capacity to increase a physiologic inflammatory response, to stimulate phagocytosis, to promote cell lysis and to enhance pathogen immunogenicity, the complement system is a crucial component of both the innate and adaptive immune responses. However, many infectious agents resist the activation of this system by expressing or secreting proteins with a role as complement regulatory, mainly inhibitory, proteins. Trypanosoma cruzi, the causal agent of Chagas disease, a reemerging microbial ailment, possesses several virulence factors with capacity to inhibit complement at different stages of activation. T. cruzi calreticulin (TcCalr) is a highly-conserved, endoplasmic reticulum-resident chaperone that the parasite translocates to the extracellular environment, where it exerts a variety of functions. Among these functions, TcCalr binds C1, MBL and ficolins, thus inhibiting the classical and lectin pathways of complement at their earliest stages of activation. Moreover, the TcCalr/C1 interaction also mediates infectivity by mimicking a strategy used by apoptotic cells for their removal. More recently, it has been determined that these Calr strategies are also used by a variety of other parasites. In addition, as reviewed elsewhere, TcCalr inhibits angiogenesis, promotes wound healing and reduces tumor growth. Complement C1 is also involved in some of these properties. Knowledge on the role of virulence factors, such as TcCalr, and their interactions with complement components in host-parasite interactions, may lead toward the description of new anti-parasite therapies and prophylaxis.


Subject(s)
Calreticulin/immunology , Complement C1/immunology , Host-Parasite Interactions/immunology , Parasites/pathogenicity , Animals , Complement Activation , Humans , Immune Evasion , Parasites/immunology , Trypanosoma cruzi/immunology , Trypanosoma cruzi/pathogenicity , Virulence Factors/immunology
16.
Mol Immunol ; 45(3): 670-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17709141

ABSTRACT

Complement is a central component of host defence, but unregulated activation can contribute to disease. The system can be initiated by three pathways: classical, alternative and lectin. The classical and lectin pathways are initiated by the C1 and mannose-binding lectin (MBL) or ficolin complexes, respectively, with C1s the executioner protease of the C1 complex and MASP-2 its counterpart in the lectin complexes. These proteases in turn cleave the C4 and C2 components of the system. Here we have elucidated the cleavage specificity of MASP-2 using a randomised substrate phage display library. Apart from the crucial P1 position, the MASP-2 S2 and S3 subsites (in that order) play the greatest role in determining specificity, with Gly residues preferred at P2 and Leu or hydrophobic residues at P3. Cleavage of peptide substrates representing the known physiological cleavage sequences in C2, C4 or the serpin C1-inhibitor (a likely regulator of MASP-2) revealed that MASP-2 is up to 1000 times more catalytically active than C1s. C1-inhibitor inhibited MASP-2 50-fold faster than C1s and much faster than any other protease tested to date, implying that MASP-2 is a major physiological target of C1-inhibitor.


Subject(s)
Complement C1 Inhibitor Protein/chemistry , Complement Pathway, Mannose-Binding Lectin/physiology , Mannose-Binding Protein-Associated Serine Proteases/chemistry , Complement C1/chemistry , Complement C1/genetics , Complement C1/immunology , Complement C1 Inhibitor Protein/genetics , Complement C1 Inhibitor Protein/immunology , Complement C2/chemistry , Complement C2/genetics , Complement C2/immunology , Complement C4/chemistry , Complement C4/genetics , Complement C4/immunology , Humans , Mannose-Binding Lectin/chemistry , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/immunology , Mannose-Binding Protein-Associated Serine Proteases/genetics , Mannose-Binding Protein-Associated Serine Proteases/immunology , Peptide Library , Substrate Specificity/physiology
17.
Front Immunol ; 10: 2537, 2019.
Article in English | MEDLINE | ID: mdl-31749804

ABSTRACT

Heterozygous missense or in-frame insertion/deletion mutations in complement 1 subunits C1r and C1s cause periodontal Ehlers-Danlos Syndrome (pEDS), a specific EDS subtype characterized by early severe periodontal destruction and connective tissue abnormalities like easy bruising, pretibial haemosiderotic plaques, and joint hypermobility. We report extensive functional studies of 16 C1R variants associated with pEDS by in-vitro overexpression studies in HEK293T cells followed by western blot, size exclusion chromatography and surface plasmon resonance analyses. Patient-derived skin fibroblasts were analyzed by western blot and Enzyme-linked Immunosorbent Assay (ELISA). Overexpression of C1R variants in HEK293T cells revealed that none of the pEDS variants was integrated into the C1 complex but cause extracellular presence of catalytic C1r/C1s activities. Variants showed domain-specific abnormalities of intracellular processing and secretion with preservation of serine protease function in the supernatant. In contrast to C1r wild type, and with the exception of a C1R missense variant disabling a C1q binding site, pEDS variants had different impact on the cell: retention of C1r fragments inside the cell, secretion of aggregates, or a new C1r cleavage site. Overexpression of C1R variants in HEK293T as well as western blot analyses of patient fibroblasts showed decreased levels of secreted C1r. Importantly, all available patient fibroblasts exhibited activated C1s and activation of externally added C4 in the supernatant while control cell lines secreted proenzyme C1s and showed no increase in C4 activation. The central elements in the pathogenesis of pEDS seem to be the intracellular activation of C1r and/or C1s, and extracellular presence of activated C1s that independently of microbial triggers can activate the classical complement cascade.


Subject(s)
Complement C1/immunology , Complement C1r/immunology , Ehlers-Danlos Syndrome/immunology , Periodontal Diseases/immunology , Cells, Cultured , Complement Activation , Complement C1r/genetics , Ehlers-Danlos Syndrome/genetics , Fibroblasts/immunology , Humans , Mutation , Periodontal Diseases/genetics
18.
J Vet Intern Med ; 33(3): 1493-1499, 2019 May.
Article in English | MEDLINE | ID: mdl-31034109

ABSTRACT

BACKGROUND: The bacterium Rhodococcus equi can cause severe pneumonia in foals. The absence of a licensed vaccine and limited effectiveness of commercial R. equi hyperimmune plasma (RE-HIP) create a great need for improved prevention of this disease. HYPOTHESIS: Plasma hyperimmune to the capsular polysaccharide poly-N-acetyl glucosamine (PNAG) would be significantly more effective than RE-HIP at mediating complement deposition and opsonophagocytic killing (OPK) of R. equi. ANIMALS: Venipuncture was performed on 9 Quarter Horses. METHODS: The ability of the following plasma sources to mediate complement component 1 (C1) deposition onto either PNAG or R. equi was determined by ELISA: (1) PNAG hyperimmune plasma (PNAG-HIP), (2) RE-HIP, and (3) standard non-hyperimmune commercial plasma (SP). For OPK, each plasma type was combined with R. equi, equine complement, and neutrophils isolated from horses (n = 9); after 4 hours, the number of R. equi in each well was determined by quantitative culture. Data were analyzed using linear mixed-effects regression with significance set at P < .05. RESULTS: The PNAG-HIP and RE-HIP were able to deposit significantly (P < .05) more complement onto their respective targets than the other plasmas. The mean proportional survival of R. equi opsonized with PNAG-HIP was significantly (P < .05) less (14.7%) than that for SP (51.1%) or RE-HIP (42.2%). CONCLUSIONS AND CLINICAL IMPORTANCE: Plasma hyperimmune to PNAG is superior to RE-HIP for opsonizing and killing R. equi in vitro. Comparison of these 2 plasmas in field trials is warranted because of the reported incomplete effectiveness of RE-HIP.


Subject(s)
Acetylglucosamine/immunology , Actinomycetales Infections/veterinary , Rhodococcus equi/immunology , Actinomycetales Infections/immunology , Animals , Antibodies, Bacterial/blood , Complement C1/immunology , Female , Horse Diseases/immunology , Horse Diseases/microbiology , Horses/immunology , Male , Neutrophils , Plasma/immunology
19.
Vaccine ; 37(9): 1142-1150, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30691984

ABSTRACT

Prescottella equi (formerly Rhodococcus equi) is a facultative intracellular bacterial pathogen that causes severe pneumonia in foals 1-6 months of age, whereas adult horses are highly resistant to infection. We have shown that vaccinating pregnant mares against the conserved surface polysaccharide capsule, ß-1 → 6-linked poly-N-acetyl glucosamine (PNAG), elicits opsonic killing antibody that transfers via colostrum to foals and protects them against experimental infection with virulent. R. equi. We hypothesized that equine IgG1 might be more important than IgG4/7 for mediating protection against R. equi infection in foals. To test this hypothesis, we compared complement component 1 (C1) deposition and polymorphonuclear cell-mediated opsonophagocytic killing (OPK) mediated by IgG1 or IgG4/7 enriched from either PNAG hyperimmune plasma (HIP) or standard plasma. Subclasses IgG1 and IgG4/7 from PNAG HIP and standard plasma were precipitated onto a diethylaminoethyl ion exchange column, then further enriched using a protein G Sepharose column. We determined C1 deposition by enzyme-linked immunosorbent assay (ELISA) and estimated OPK by quantitative microbiologic culture. Anti-PNAG IgG1 deposited significantly (P < 0.05) more C1 onto PNAG than did IgG4/7 from PNAG HIP or subclasses IgG1 and IgG4/7 from standard plasma. In addition, IgG1 from PNAG HIP mediated significantly (P < 0.05) greater OPK than IgG4/7 from PNAG HIP or IgG1 and IgG4/7 from standard plasma. Our findings indicate that anti-PNAG IgG1 is a correlate of protection against R. equi in foals, which has important implications for understanding the immunopathogenesis of R. equi pneumonia, and as a tool for assessing vaccine efficacy and effectiveness when challenge is not feasible.


Subject(s)
Acetylglucosamine/immunology , Actinomycetales Infections/veterinary , Antibodies, Bacterial/blood , Complement C1/immunology , Immunoglobulin G/blood , Phagocytosis , Rhodococcus equi/immunology , Actinomycetales Infections/immunology , Actinomycetales Infections/prevention & control , Age Factors , Animals , Animals, Newborn , Antibodies, Bacterial/classification , Antibodies, Bacterial/immunology , Horse Diseases/immunology , Horse Diseases/prevention & control , Horses/immunology , Immunoglobulin G/classification , Opsonin Proteins , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/prevention & control
20.
Adv Exp Med Biol ; 632: 237-51, 2008.
Article in English | MEDLINE | ID: mdl-19025126

ABSTRACT

C1 is a multimolecular complex that initiates the classical pathway of complement. It is composed of the pattern recognition component C1q and the serine proteases C1r and C1s. Activation of C1 elicits a series of potent effector mechanisms directed at limiting infection by invading pathogens as well as participating in other biological functions such as immune tolerance. While many molecules in addition to antibody have been demonstrated to activate C1, only a handful of C1 inhibitors have been described. Disregulated control of complement activation is associated with numerous autoimmune and inflammatory disease processes, thus tight regulation of C1 activation is highly desirable. We have recently discovered a novel inhibitor of C1, the coat protein of the human astroviruses, a family of enteric pathogens that infect young children. The astrovirus coat protein binds to the A-chain of C1q and inhibits spontaneous as well as antibody-mediated activation of the C1 complex resulting in suppression of classical pathway activation and complement-mediated terminal effector functions. This is the first description of a non-enveloped icosahedral virus inhibiting complement activation and the first description of a viral inhibitor of C1. The known inhibitors of C1 are reviewed and then discussed in the context of this novel viral C1 inhibitor. Additionally, the properties of this compound are elucidated highlighting its potential as an anti-complement therapeutic for the many diseases associated with inappropriate complement activation.


Subject(s)
Capsid Proteins/immunology , Complement Activation/immunology , Complement C1 Inhibitor Protein/immunology , Complement C1/antagonists & inhibitors , Mamastrovirus/immunology , Complement C1/immunology , Complement C1q/immunology , Complement C1q/metabolism , Complement C1r/immunology , Complement C1r/metabolism , Complement C1s/immunology , Complement C1s/metabolism , Humans , Models, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL