Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1151731, 2023.
Article in English | MEDLINE | ID: mdl-37180096

ABSTRACT

Complement C1s association with the pathogenesis of several diseases cannot be simply explained only by considering its main role in activating the classical complement pathway. This suggests that non-canonical functions are to be deciphered for this protease. Here the focus is on C1s cleavage of HMGB1 as an auxiliary target. HMGB1 is a chromatin non-histone nuclear protein, which exerts in fact multiple functions depending on its location and its post-translational modifications. In the extracellular compartment, HMGB1 can amplify immune and inflammatory responses to danger associated molecular patterns, in health and disease. Among possible regulatory mechanisms, proteolytic processing could be highly relevant for HMGB1 functional modulation. The unique properties of HMGB1 cleavage by C1s are analyzed in details. For example, C1s cannot cleave the HMGB1 A-box fragment, which has been described in the literature as an inhibitor/antagonist of HMGB1. By mass spectrometry, C1s cleavage was experimentally identified to occur after lysine on position 65, 128 and 172 in HMGB1. Compared to previously identified C1s cleavage sites, the ones identified here are uncommon, and their analysis suggests that local conformational changes are required before cleavage at certain positions. This is in line with the observation that HMGB1 cleavage by C1s is far slower when compared to human neutrophil elastase. Recombinant expression of cleavage fragments and site-directed mutagenesis were used to confirm these results and to explore how the output of C1s cleavage on HMGB1 is finely modulated by the molecular environment. Furthermore, knowing the antagonist effect of the isolated recombinant A-box subdomain in several pathophysiological contexts, we wondered if C1s cleavage could generate natural antagonist fragments. As a functional readout, IL-6 secretion following moderate LPS activation of RAW264.7 macrophage was investigated, using LPS alone or in complex with HMGB1 or some recombinant fragments. This study revealed that a N-terminal fragment released by C1s cleavage bears stronger antagonist properties as compared to the A-box, which was not expected. We discuss how this fragment could provide a potent brake for the inflammatory process, opening the way to dampen inflammation.


Subject(s)
Complement C1s , HMGB1 Protein , Humans , Complement C4/metabolism , Lipopolysaccharides , Anti-Inflammatory Agents
2.
Front Immunol ; 13: 869720, 2022.
Article in English | MEDLINE | ID: mdl-35572583

ABSTRACT

Our immune system responds to infectious (PAMPs) and tissue damage (DAMPs) signals. The complement system and alarmin High-Mobility Group Box 1 (HMGB1) are two powerful soluble actors of human host defense and immune surveillance. These systems involve molecular cascades and amplification loops for their signaling or activation. Initially activated as alarm raising systems, their function can be finally switched towards inflammation resolution, where they sustain immune maturation and orchestrate repair mechanisms, opening the way back to homeostasis. However, when getting out of control, these defense systems can become deleterious and trigger serious cellular and tissue damage. Therefore, they can be considered as double-edged swords. The close interaction between the complement and HMGB1 pathways is described here, as well as their traditional and non-canonical roles, their functioning at different locations and their independent and collective impact in different systems both in health and disease. Starting from these systems and interplay at the molecular level (when elucidated), we then provide disease examples to better illustrate the signs and consequences of their roles and interaction, highlighting their importance and possible vicious circles in alarm raising and inflammation, both individually or in combination. Although this integrated view may open new therapeutic strategies, future challenges have to be faced because of the remaining unknowns regarding the molecular mechanisms underlying the fragile molecular balance which can drift towards disease or return to homeostasis, as briefly discussed at the end.


Subject(s)
HMGB1 Protein , Alarmins , Complement System Proteins , HMGB1 Protein/metabolism , Humans , Inflammation , Signal Transduction/physiology
3.
Int J Mol Sci ; 22(10)2021 May 12.
Article in English | MEDLINE | ID: mdl-34066122

ABSTRACT

The immune system homeostasis relies on a tight equilibrium of interconnected stimulatory and inhibitory signals. Disruption of this balance is characteristic of autoimmune diseases such as systemic lupus erythematosus (SLE). Aside from activating the classical complement pathway and enhancing pathogens and apoptotic cells phagocytosis, C1q has been recently shown to play an important role in immune modulation and tolerance by interacting with several inhibitory and stimulatory immune receptors. Due to its functional organization into collagen-like (CLR) and globular (GR) regions and its multimeric nature, C1q is able to interact simultaneously with several of these receptors and locally congregate pro- and anti-inflammatory signals, thus modulating the immune response. Leukocyte associated immunoglobulin-like (Ig-like) receptor 1 (LAIR-1), a ubiquitous collagen receptor expressed in many immune cell types, has been reported to interact with the CLR of C1q. In this study, we provide new insights into the molecular and structural determinants underlying C1q/LAIR-1 interaction. Recombinant LAIR-1 extracellular Ig-like domain was produced and tested for its interaction with C1q. A molecular dissection of C1q combined with competition assays reveals that LAIR-1 interacts with C1q's CLR through a binding site close but different from the one of its associated C1r2s2 proteases tetramer. On the other side, we identified LAIR-1 residues involved in C1q interaction by site-directed mutational analysis. All together, these results lead to propose a possible model for C1q interaction with LAIR-1 and will contribute to the fundamental understanding of C1q-mediated immune tolerance.


Subject(s)
Collagen/metabolism , Complement C1q/metabolism , Receptors, Immunologic/metabolism , Binding Sites , Complement C1q/genetics , Humans , Immune Tolerance , Mutation , Protein Binding , Receptors, Immunologic/genetics
4.
J Immunol Methods ; 492: 113001, 2021 05.
Article in English | MEDLINE | ID: mdl-33621564

ABSTRACT

Complement C1q is a multifunctional protein able to sense pathogens and immune molecules such as immunoglobulins and pentraxins, and to trigger the classical complement pathway through activation of its two associated proteases, C1r and C1s. C1q is a multimeric protein composed of three homologous yet distinct polypeptide chains A, B, and C, each composed of an N-terminal collagen-like sequence and a C-terminal globular gC1q module, that assemble into six heterotrimeric (A-B-C) subunits. This hexameric structure exhibits the characteristic shape of a bouquet of flowers, comprising six collagen-like triple helices, each terminating in a trimeric C-terminal globular head. We have produced previously functional recombinant full-length C1q in stably transfected HEK 293-F cells, with a FLAG tag inserted at the C-terminal end of C1qC chain. We report here the generation of additional recombinant C1q proteins, with a FLAG tag fused to the C-terminus of C1qA or C1qB chains, or to the N-terminus of the C1qC chain. Two other variants harboring a Myc or a 6-His tag at the C-terminal end of C1qC were also produced. We show that all C1q variants, except for the His-tagged protein, can be produced at comparable yields and are able to bind with similar affinities to either IgM, a ligand of the globular regions, or to the C1r2-C1s2 tetramer, and to trigger IgM-mediated serum complement activation. These new recombinant C1q variants provide additional tools to investigate the multiple functions of C1q.


Subject(s)
Complement C1q/isolation & purification , Molecular Probes/genetics , Amino Acid Sequence , Complement Activation , Complement C1q/genetics , Complement C1q/metabolism , HEK293 Cells , Humans , Immunoassay/methods , Protein Multimerization , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Transfection
5.
FEBS J ; 288(6): 2030-2041, 2021 03.
Article in English | MEDLINE | ID: mdl-32869492

ABSTRACT

Complement component C1q, a soluble defense collagen, is the recognition protein of the classical complement pathway. C1q is able to recognize and interact with multiple targets and, via the subsequent activation of its cognate serine proteases C1r and C1s, initiates the complement cascade. C1q is made up of six ABC heterotrimers each containing two different functional regions, an N-terminal collagen-like region (CLR) and a C-terminal globular region (GR). These heterotrimers assemble via their N-terminal regions, resulting in the characteristic 'bouquet-like' shape of C1q with an N-terminal bundle of collagen fibers with six diverging stems each exhibiting a C-terminal globular head. The GRs are responsible for the versatile recognition of multiple C1q targets, whereas the CLRs trigger immune response through interacting with several cellular or soluble partners. We report here the generation of the first recombinant form of human C1q without its recognition globular heads. The noncollagenous domain 2 (nc2) of type IX collagen has been substituted for the C1q GR in order to control the correct registering of the collagen triple helices of C1q chains A, B, and C. The resulting CLR_nc2 recombinant protein produced in stably transfected EXPI293 mammalian cells was correctly assembled and folded, as demonstrated by mass spectrometry, mass photometry, and electron microscopy experiments. Its interaction properties were investigated using surface plasmon resonance analysis with known CLR ligands: the tetramer of C1r and C1s dimers and MBL-associated protein MAp44. Comparison with the interaction properties of native serum-derived C1q and CLR revealed that recombinant CLR_nc2 retains C1q CLR functional properties.


Subject(s)
Complement C1q/chemistry , Protein Domains , Protein Multimerization , Recombinant Proteins/chemistry , Amino Acid Sequence , Collagen/chemistry , Collagen/genetics , Collagen/metabolism , Complement Activation/genetics , Complement C1q/genetics , Complement C1q/metabolism , Humans , Ligands , Mass Spectrometry , Microscopy, Electron , Mutation, Missense , Photometry , Protein Binding , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Surface Plasmon Resonance
6.
Front Immunol ; 11: 583754, 2020.
Article in English | MEDLINE | ID: mdl-33193398

ABSTRACT

LRP1 is a large endocytic modular receptor that plays a crucial role in the scavenging of apoptotic material through binding to pattern-recognition molecules. It is a membrane anchored receptor of the LDL receptor family with 4 extracellular clusters of ligand binding modules called cysteine rich complement-type repeats that are involved in the interaction of LRP1 with its numerous ligands. Complement C1q was shown to interact with LRP1 and to be implicated in the phagocytosis of apoptotic cells. The present work aimed at exploring how these two large molecules interact at the molecular level using a dissection strategy. For that purpose, recombinant LRP1 clusters II, III and IV were produced in mammalian HEK293F cells and their binding properties were investigated. Clusters II and IV were found to interact specifically and efficiently with C1q with K Ds in the nanomolar range. The use of truncated C1q fragments and recombinant mutated C1q allowed to localize more precisely the binding site for LRP1 on the collagen-like regions of C1q (CLRs), nearby the site that is implicated in the interaction with the cognate protease tetramer C1r2s2. This site could be a common anchorage for other ligands of C1q CLRs such as sulfated proteoglycans and Complement receptor type 1. The use of a cellular model, consisting in CHO LRP1-null cells transfected with full-length LRP1 or a cluster IV minireceptor (mini IV) confirmed that mini IV interacts with C1q at the cell membrane as well as full-length LRP1. Further cellular interaction studies finally highlighted that mini IV can endorse the full-length LRP1 binding efficiency for apoptotic cells and that C1q has no impact on this interaction.


Subject(s)
Complement C1q/metabolism , Complement C1r/metabolism , Complement C1s/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Peptide Hydrolases/metabolism , Animals , Apoptosis/physiology , Binding Sites/physiology , CHO Cells , Cell Line , Cell Membrane/metabolism , Cricetulus , HEK293 Cells , Humans , Ligands , Protein Domains/physiology
8.
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
9.
Front Immunol ; 10: 2962, 2019.
Article in English | MEDLINE | ID: mdl-31921203

ABSTRACT

Ehlers-Danlos syndromes (EDS) are clinically and genetically heterogeneous disorders characterized by soft connective tissue alteration like joint hypermobility and skin hyper-extensibility. We previously identified heterozygous missense mutations in the C1R and C1S genes, coding for the complement C1 proteases, in patients affected by periodontal EDS, a specific EDS subtype hallmarked by early severe periodontitis leading to premature loss of teeth and connective tissue alterations. Up to now, there is no clear molecular link relating the nominal role of the C1r and C1s proteases, which is to activate the classical complement pathway, to these heterogeneous symptoms of periodontal EDS syndrome. We aim therefore to elucidate the functional effect of these mutations, at the molecular and enzymatic levels. To explore the molecular consequences, a set of cell transfection experiments, recombinant protein purification, mass spectroscopy and N-terminal analyses have been performed. Focusing on the results obtained on two different C1S variants, namely p.Val316del and p.Cys294Arg, we show that HEK293-F cells stably transfected with the corresponding C1s variant plasmids, unexpectedly, do not secrete the full-length mutated C1s, but only a truncated Fg40 fragment of 40 kDa, produced at very low levels. Detailed analyses of the Fg40 fragments purified for the two C1s variants show that they are identical, which was also unexpected. This suggests that local misfolding of the CCP1 module containing the patient mutation exposes a novel cleavage site, between Lys353 and Cys354, which is not normally accessible. The mutation-induced Fg40 fragment contains the intact C-terminal serine protease domain but not the N-terminal domain mediating C1s interaction with the other C1 subunits, C1r, and C1q. Thus, Fg40 enzymatic activity escapes the normal physiological control of C1s activity within C1, potentially providing a loss-of-control. Comparative enzymatic analyses show that Fg40 retains the native esterolytic activity of C1s, as well as its cleavage efficiency toward the ancillary alarmin HMGB1 substrate, for example, whereas the nominal complement C4 activation cleavage is impaired. These new results open the way to further molecular explorations possibly involving subsidiary C1s targets.


Subject(s)
Complement C1r , Complement C1s , Ehlers-Danlos Syndrome , Mutation, Missense , Periodontal Diseases , Amino Acid Substitution , Complement C1r/genetics , Complement C1r/immunology , Complement C1s/genetics , Complement C1s/immunology , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/immunology , Ehlers-Danlos Syndrome/pathology , HEK293 Cells , Humans , Periodontal Diseases/genetics , Periodontal Diseases/immunology , Periodontal Diseases/pathology , Protein Folding
10.
Front Immunol ; 9: 453, 2018.
Article in English | MEDLINE | ID: mdl-29563915

ABSTRACT

Complement receptor type 1 (CR1) is a multi modular membrane receptor composed of 30 homologous complement control protein modules (CCP) organized in four different functional regions called long homologous repeats (LHR A, B, C, and D). CR1 is a receptor for complement-opsonins C3b and C4b and specifically interacts through pairs of CCP modules located in LHR A, B, and C. Defense collagens such as mannose-binding lectin (MBL), ficolin-2, and C1q also act as opsonins and are involved in immune clearance through binding to the LHR-D region of CR1. Our previous results using deletion variants of CR1 mapped the interaction site for MBL and ficolin-2 on CCP24-25. The present work aimed at deciphering the interaction of C1q with CR1 using new CR1 variants concentrated around CCP24-25. CR1 bimodular fragment CCP24-25 and CR1 CCP22-30 deleted from CCP24-25 produced in eukaryotic cells enabled to highlight that the interaction site for both MBL and C1q is located on the same pair of modules CCP24-25. C1q binding to CR1 shares with MBL a main common interaction site on the collagen stalks but also subsidiary sites most probably located on C1q globular heads, contrarily to MBL.


Subject(s)
Complement C1q/chemistry , Mannose-Binding Lectin/chemistry , Peptides/chemistry , Receptors, Complement 3b/chemistry , Complement C1q/genetics , Complement C1q/immunology , Humans , Lectins/chemistry , Lectins/genetics , Lectins/immunology , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/immunology , Peptides/genetics , Peptides/immunology , Protein Binding , Protein Domains , Protein Structure, Secondary , Receptors, Complement 3b/genetics , Receptors, Complement 3b/immunology , Ficolins
11.
Front Immunol ; 5: 565, 2014.
Article in English | MEDLINE | ID: mdl-25414705

ABSTRACT

The classical complement pathway is initiated by the large (~800 kDa) and flexible multimeric C1 complex. Its catalytic function is triggered by the proteases hetero-tetramer C1r2s2, which is associated to the C1q sensing unit, a complex assembly of 18 chains built as a hexamer of heterotrimers. Initial pioneering studies gained insights into the main architectural principles of the C1 complex. A dissection strategy then provided the high-resolution structures of its main functional and/or structural building blocks, as well as structural details on some key protein-protein interactions. These past and current discoveries will be briefly summed up in order to address the question of what is still ill-defined. On a functional point of view, the main molecular determinants of C1 activation and its tight control will be delineated. The current perspective remains to decipher how C1 really works and is controlled in vivo, both in normal and pathological settings.

12.
Methods Mol Biol ; 1100: 43-60, 2014.
Article in English | MEDLINE | ID: mdl-24218249

ABSTRACT

C1r and C1s are the proteases responsible for the activation and proteolytic activity of the C1 complex of the classical complement pathway, respectively. They are assembled into a Ca(2+)-dependent C1s-C1r-C1r-C1s tetramer which in turn associates with the recognition protein C1q. The C1 complex circulates in serum as a zymogen and is activated upon binding of C1q to appropriate targets, such as antigen-antibody complexes. This property is used for the purification of C1r and C1s from human serum after binding of C1 to insoluble immune complexes. Disruption of the bound C1 complex by EDTA releases C1r and C1s which are further separated by ion-exchange chromatography; both proteins can be reassembled in the presence of calcium ions and the reconstituted tetramer isolated by gel filtration. In this chapter, we describe the purification of the activated and proenzyme forms of C1r and C1s and of the proenzyme C1s-C1r-C1r-C1s tetramer as well as methods for their biochemical and functional characterization. The production of recombinant C1s and of the proenzyme tetramer in a baculovirus-insect cell system, and their purification by affinity chromatography is also presented.


Subject(s)
Complement C1r/immunology , Complement C1s/immunology , Complement Pathway, Classical , Animals , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/isolation & purification , Antigen-Antibody Complex/metabolism , Complement C1r/isolation & purification , Complement C1s/isolation & purification , Gene Expression , Humans , Protein Binding/immunology , Protein Multimerization , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Sf9 Cells , Solubility
13.
PLoS One ; 8(7): e67962, 2013.
Article in English | MEDLINE | ID: mdl-23861840

ABSTRACT

Mannan-binding lectin (MBL), ficolins and collectin-11 are known to associate with three homologous modular proteases, the MBL-Associated Serine Proteases (MASPs). The crystal structures of the catalytic domains of MASP-1 and MASP-2 have been solved, but the structure of the corresponding domain of MASP-3 remains unknown. A link between mutations in the MASP1/3 gene and the rare autosomal recessive 3MC (Mingarelli, Malpuech, Michels and Carnevale,) syndrome, characterized by various developmental disorders, was discovered recently, revealing an unexpected important role of MASP-3 in early developmental processes. To gain a first insight into the enzymatic and structural properties of MASP-3, a recombinant form of its serine protease (SP) domain was produced and characterized. The amidolytic activity of this domain on fluorescent peptidyl-aminomethylcoumarin substrates was shown to be considerably lower than that of other members of the C1r/C1s/MASP family. The E. coli protease inhibitor ecotin bound to the SP domains of MASP-3 and MASP-2, whereas no significant interaction was detected with MASP-1, C1r and C1s. A tetrameric complex comprising an ecotin dimer and two MASP-3 SP domains was isolated and its crystal structure was solved and refined to 3.2 Å. Analysis of the ecotin/MASP-3 interfaces allows a better understanding of the differential reactivity of the C1r/C1s/MASP protease family members towards ecotin, and comparison of the MASP-3 SP domain structure with those of other trypsin-like proteases yields novel hypotheses accounting for its zymogen-like properties in vitro.


Subject(s)
Escherichia coli Proteins/chemistry , Mannose-Binding Protein-Associated Serine Proteases/chemistry , Molecular Docking Simulation , Periplasmic Proteins/chemistry , Binding Sites , Catalytic Domain , Complement C1r/chemistry , Complement C1s/chemistry , Coumarins , Crystallography, X-Ray , Enzyme Assays , Fluorescent Dyes , Humans , Mannose-Binding Protein-Associated Serine Proteases/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Substrate Specificity , Thrombin/chemistry , Trypsin/chemistry
14.
J Immunol ; 190(7): 3721-31, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23460739

ABSTRACT

Complement receptor type 1 (CR1) is a membrane receptor expressed on a wide range of cells. It is involved in immune complex clearance, phagocytosis, and complement regulation. Its ectodomain is composed of 30 complement control protein (CCP) modules, organized into four long homologous repeats (A-D). In addition to its main ligands C3b and C4b, CR1 was reported to interact with C1q and mannan-binding lectin (MBL) likely through its C-terminal region (CCP22-30). To decipher the interaction of human CR1 with the recognition proteins of the lectin complement pathway, a recombinant fragment encompassing CCP22-30 was expressed in eukaryotic cells, and its interaction with human MBL and ficolins was investigated using surface plasmon resonance spectroscopy. MBL and L-ficolin were shown to interact with immobilized soluble CR1 and CR1 CCP22-30 with apparent dissociation constants in the nanomolar range, indicative of high affinity. The binding site for CR1 was located at or near the MBL-associated serine protease (MASP) binding site in the collagen stalks of MBL and L-ficolin, as shown by competition experiments with MASP-3. Accordingly, the mutation of an MBL conserved lysine residue essential for MASP binding (K55) abolished binding to soluble CR1 and CCP22-30. The CR1 binding site for MBL/ficolins was mapped to CCP24-25 of long homologous repeat D using deletion mutants. In conclusion, we show that ficolins are new CR1 ligands and propose that MBL/L-ficolin binding involves major ionic interactions between conserved lysine residues of their collagen stalks and surface exposed acidic residues located in CR1 CCP24 and/or CCP25.


Subject(s)
Complement Pathway, Mannose-Binding Lectin , Mannose-Binding Lectin/metabolism , Receptors, Complement/metabolism , Binding Sites , Carrier Proteins/metabolism , Complement Pathway, Mannose-Binding Lectin/genetics , Humans , Kinetics , Lectins/metabolism , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Receptors, Complement/chemistry , Receptors, Complement/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ficolins
15.
J Immunol ; 184(9): 4982-9, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20351192

ABSTRACT

Variants of the human C1 inhibitor serpin domain containing three N-linked carbohydrates at positions 216, 231, and 330 (C1inhDelta97), a single carbohydrate at position 330 (C1inhDelta97DM), or no carbohydrate were produced in a baculovirus/insect cells system. An N-terminally His-tagged C1inhDelta97 variant was also produced. Removal of the oligosaccharide at position 330 dramatically decreased expression, precluding further analysis. All other variants were characterized chemically and shown to inhibit C1s activity and C1 activation in the same way as native C1 inhibitor. Likewise, they formed covalent complexes with C1s as shown by SDS-PAGE analysis. C1 inhibitor and its variants inhibited the ability of C1r-like protease to activate C1s, but did not form covalent complexes with this protease. The interaction of C1 inhibitor and its variants with heparin was investigated by surface plasmon resonance, yielding K(D) values of 16.7 x 10(-8) M (C1 inhibitor), 2.3 x 10(-8) M (C1inhDelta97), and 3.6 x 10(-8) M (C1inhDelta97DM). C1s also bound to heparin, with lower affinity (K(D) = 108 x 10(-8) M). Using the same technique, 50% inhibition of the binding of C1 inhibitor and C1s to heparin was achieved using heparin oligomers containing eight and six saccharide units, respectively. These values roughly correlate with the size of 10 saccharide units yielding half-maximal potentiation of the inhibition of C1s activity by C1 inhibitor, consistent with a "sandwich" mechanism. Using a thermal shift assay, heparin was shown to interact with the C1s serine protease domain and the C1 inhibitor serpin domain, increasing and decreasing their thermal stability, respectively.


Subject(s)
Complement C1 Inhibitor Protein/physiology , Complement C1/antagonists & inhibitors , Complement C1/metabolism , Heparin/metabolism , Serpins/physiology , Animals , Baculoviridae/genetics , Binding, Competitive/genetics , Binding, Competitive/immunology , Carbohydrates/chemistry , Carbohydrates/genetics , Complement C1 Inhibitor Protein/genetics , Complement C1 Inhibitor Protein/metabolism , Complement C1s/antagonists & inhibitors , Complement C1s/metabolism , Heparin/chemistry , Humans , Molecular Weight , Moths/genetics , Mutagenesis, Site-Directed , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Serpins/genetics , Serpins/metabolism , Spodoptera/genetics , Swine
16.
Mol Immunol ; 47(7-8): 1553-60, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20153530

ABSTRACT

The two N-linked oligosaccharides in native human C9 were deleted by site-specific mutagenesis. This aglycosyl-C9 did not differ from its native form in hemolytic and bactericidal activity. A new N-glycosylation site (K311N/E313T) was introduced into the turn of a helix-turn-helix [HTH] fold that had been postulated to form a transmembrane hairpin in membrane-bound C9. This glycosylated form of human C9 was as active as the native protein suggesting that the glycan chain remains on the external side of the membrane and that translocation of this hairpin is not required for membrane anchoring. Furthermore, flow cytometry provided evidence for the recognition of membrane-bound C9 on complement-lysed ghosts by an antibody specific for the HTH fold. A new N-glycosylation site (P26N) was also introduced close to the N-terminus of C9 to test whether this region was involved in C9 polymerization, which is thought to be required for cytolytic activity of C9. Again, this glycosylated C9 was as active as native C9 and could be induced to polymerize by heating or incubation with metal ions. The two C-terminal cystines within the MACPF domain could be eliminated partially or completely without affecting the hemolytic activity. Free sulfhydryl groups of unpaired cysteines in such C9 mutants are blocked since they could not be modified with SH-specific reagents. These results are discussed with respect to a recently proposed model that, on the basis of the MACPF structure in C8alpha, envisions membrane insertion of C9 to resemble the mechanism by which cholesterol-dependent cytolysins enter a membrane.


Subject(s)
Antibodies/immunology , Cell Membrane/chemistry , Cell Membrane/metabolism , Complement C9/chemistry , Complement C9/metabolism , Disulfides/metabolism , Peptides/immunology , Amino Acid Sequence , Animals , Cell Membrane/immunology , Chlorocebus aethiops , Complement C9/genetics , Complement C9/immunology , Glycosylation , Humans , Molecular Sequence Data , Mutation , Peptide Mapping , Protein Folding , Protein Structure, Secondary , Sequence Alignment , Spodoptera
17.
J Biol Chem ; 284(29): 19340-8, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19473974

ABSTRACT

The C1 complex of complement is assembled from a recognition protein C1q and C1s-C1r-C1r-C1s, a Ca(2+)-dependent tetramer of two modular proteases C1r and C1s. Resolution of the x-ray structure of the N-terminal CUB(1)-epidermal growth factor (EGF) C1s segment has led to a model of the C1q/C1s-C1r-C1r-C1s interaction where the C1q collagen stem binds at the C1r/C1s interface through ionic bonds involving acidic residues contributed by the C1r EGF module (Gregory, L. A., Thielens, N. M., Arlaud, G. J., Fontecilla-Camps, J. C., and Gaboriaud, C. (2003) J. Biol. Chem. 278, 32157-32164). To identify the C1q-binding sites of C1s-C1r-C1r-C1s, a series of C1r and C1s mutants was expressed, and the C1q binding ability of the resulting tetramer variants was assessed by surface plasmon resonance. Mutations targeting the Glu(137)-Glu-Asp(139) stretch in the C1r EGF module had no effect on C1 assembly, ruling out our previous interaction model. Additional mutations targeting residues expected to participate in the Ca(2+)-binding sites of the C1r and C1s CUB modules provided evidence for high affinity C1q-binding sites contributed by the C1r CUB(1) and CUB(2) modules and lower affinity sites contributed by C1s CUB(1). All of the sites implicate acidic residues also contributing Ca(2+) ligands. C1s-C1r-C1r-C1s thus contributes six C1q-binding sites, one per C1q stem. Based on the location of these sites and available structural information, we propose a refined model of C1 assembly where the CUB(1)-EGF-CUB(2) interaction domains of C1r and C1s are entirely clustered inside C1q and interact through six binding sites with reactive lysines of the C1q stems. This mechanism is similar to that demonstrated for mannan-binding lectin (MBL)-MBL-associated serine protease and ficolin-MBL-associated serine protease complexes.


Subject(s)
Complement C1 Inactivator Proteins/metabolism , Complement C1q/metabolism , Complement C1r/metabolism , Amino Acid Sequence , Binding Sites/genetics , Calcium/metabolism , Complement C1 Inactivator Proteins/chemistry , Complement C1 Inactivator Proteins/genetics , Complement C1 Inhibitor Protein , Complement C1q/chemistry , Complement C1q/genetics , Complement C1r/chemistry , Complement C1r/genetics , Electrophoresis, Polyacrylamide Gel , Epidermal Growth Factor/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Mutation , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Surface Plasmon Resonance
18.
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
19.
J Biol Chem ; 280(51): 41811-8, 2005 Dec 23.
Article in English | MEDLINE | ID: mdl-16227207

ABSTRACT

C1s and mannan-binding lectin-associated serine protease-2 (MASP-2) are the proteases that trigger the classical and lectin pathways of complement, respectively. They have identical modular architectures and cleave the same substrates, C2 and C4, but show markedly different efficiencies toward C4. Multisite-directed mutagenesis was used to engineer hybrid C1s/MASP-2 molecules where either the complement control protein (CCP) modules or the serine protease (SP) domain of C1s were swapped for their MASP-2 counterparts. The resulting chimeras (C1s(MASP-2 CCP1/2) and C1s(MASP-2 SP), respectively) were expressed and characterized chemically and functionally. Whereas C1s(MASP-2 SP) was recovered as an active enzyme, C1s(MASP-2 CCP1/2) was produced in a proenzyme form and was susceptible to activation by C1r, indicating that the activation properties of the chimeras were dictated by the nature of their SP domain. Similarly, each activated chimera had an esterolytic activity characteristic of its own SP domain and cleaved C2 with an efficiency comparable with that of their parent C1s and MASP-2 proteases. Both chimeras cleaved C4, but whereas C1s(MASP-2 SP) and C1s had Km values in the micromolar range, C1s(MASP-2 CCP1/2) and MASP-2 had Km values in the nanomolar range, resulting in 21-27-fold higher kcat/Km ratios. Thus, the higher C4 cleavage efficiency of MASP-2 arises from a higher substrate recognition efficacy of its CCP modules. Remarkably, C1s(MASP-2 CCP1/2) retained C1s ability to associate with C1r and C1q to form a pseudo-C1 complex and to undergo activation within this complex, indicating that the C1s-CCP modules have no direct implication in either function.


Subject(s)
Mannose-Binding Protein-Associated Serine Proteases/metabolism , Recombinant Fusion Proteins/metabolism , Amino Acid Sequence , Animals , Electrophoresis, Polyacrylamide Gel , Protein Binding
20.
J Immunol ; 175(7): 4536-42, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16177097

ABSTRACT

C1s is the modular serine protease responsible for cleavage of C4 and C2, the protein substrates of the first component of C (C1). Its catalytic domain comprises two complement control protein (CCP) modules connected by a four-residue linker Gln340-Pro-Val-Asp343 and a serine protease domain. To assess the functional role of the linker, a series of mutations were performed at positions 340-343 of human C1s, and the resulting mutants were produced using a baculovirus-mediated expression system and characterized functionally. All mutants were secreted in a proenzyme form and had a mass of 77,203-77,716 Da comparable to that of wild-type C1s, except Q340E, which had a mass of 82,008 Da, due to overglycosylation at Asn391. None of the mutations significantly altered C1s ability to assemble with C1r and C1q within C1. Whereas the other mutations had no effect on C1s activation, the Q340E mutant was totally resistant to C1r-mediated activation, both in the fluid phase and within the C1 complex. Once activated, all mutants cleaved C2 with an efficiency comparable to that of wild-type C1s. In contrast, most of the mutations resulted in a decreased C4-cleaving activity, with particularly pronounced inhibitory effects for point mutants Q340K, P341I, V342K, and D343N. Comparable effects were observed when the C4-cleaving activity of the mutants was measured inside C1. Thus, flexibility of the C1s CCP1-CCP2 linker plays no significant role in C1 assembly or C1s activation by C1r inside C1 but plays a critical role in C4 cleavage by adjusting positioning of this substrate for optimal cleavage by the C1s active site.


Subject(s)
Complement C1s/physiology , Amino Acid Motifs , Amino Acid Substitution/genetics , Animals , Asparagine/metabolism , Baculoviridae , Complement C1s/genetics , Complement C2/metabolism , Complement C4/metabolism , Genetic Vectors , Glycosylation , Humans , Kinetics , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Spodoptera
SELECTION OF CITATIONS
SEARCH DETAIL
...