Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Biomolecules ; 11(2)2021 02 17.
Article in English | MEDLINE | ID: mdl-33671302

ABSTRACT

The complement system is part of the innate immune response, where it provides immediate protection from infectious agents and plays a fundamental role in homeostasis. Complement dysregulation occurs in several diseases, where the tightly regulated proteolytic cascade turns offensive. Prominent examples are atypical hemolytic uremic syndrome, paroxysmal nocturnal hemoglobinuria and Alzheimer's disease. Therapeutic intervention targeting complement activation may allow treatment of such debilitating diseases. In this review, we describe a panel of complement targeting nanobodies that allow modulation at different steps of the proteolytic cascade, from the activation of the C1 complex in the classical pathway to formation of the C5 convertase in the terminal pathway. Thorough structural and functional characterization has provided a deep mechanistic understanding of the mode of inhibition for each of the nanobodies. These complement specific nanobodies are novel powerful probes for basic research and offer new opportunities for in vivo complement modulation.


Subject(s)
Complement Activation , Complement System Proteins , Nanomedicine/methods , Single-Domain Antibodies/chemistry , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/drug therapy , Atypical Hemolytic Uremic Syndrome/immunology , Complement C1/chemistry , Complement C3-C5 Convertases/chemistry , Epitopes/chemistry , Hemoglobinuria, Paroxysmal/drug therapy , Hemoglobinuria, Paroxysmal/immunology , Humans , Immunity, Innate , Immunoglobulin G/immunology , Inflammation , Molecular Conformation , Protein Binding , Proteolysis
2.
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
3.
Science ; 359(6377): 794-797, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29449492

ABSTRACT

Danger patterns on microbes or damaged host cells bind and activate C1, inducing innate immune responses and clearance through the complement cascade. How these patterns trigger complement initiation remains elusive. Here, we present cryo-electron microscopy analyses of C1 bound to monoclonal antibodies in which we observed heterogeneous structures of single and clustered C1-immunoglobulin G1 (IgG1) hexamer complexes. Distinct C1q binding sites are observed on the two Fc-CH2 domains of each IgG molecule. These are consistent with known interactions and also reveal additional interactions, which are supported by functional IgG1-mutant analysis. Upon antibody binding, the C1q arms condense, inducing rearrangements of the C1r2s2 proteases and tilting C1q's cone-shaped stalk. The data suggest that C1r may activate C1s within single, strained C1 complexes or between neighboring C1 complexes on surfaces.


Subject(s)
Alarmins/chemistry , Complement Activation , Complement C1/chemistry , Immunoglobulin G/chemistry , Alarmins/ultrastructure , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/ultrastructure , Binding Sites , Complement C1/ultrastructure , Cryoelectron Microscopy , Humans , Immunoglobulin G/genetics , Immunoglobulin G/ultrastructure
4.
Protein Sci ; 25(10): 1883-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27391278

ABSTRACT

C1q contains three globular domains (C1qgD) that are the key functional component of the classical complement system. C1qgD can interact with important immune molecules, including IgG and C-reactive protein (CRP) to form defense systems to protect animals. Here, the first non-mammalian structure, zebrafish C1qA globular domain (Dare-C1qAgD) was solved. Although the overall architecture of Dare-C1qAgD is similar to human C1qA, residues involved in C1qBgD, C1qCgD, and CRP binding are somewhat different while residues involved in IgG binding are not present in zebrafish. The structure gives insight into how human and fish C1qA evolved from an ancestral protein.


Subject(s)
Complement C1/chemistry , Evolution, Molecular , Zebrafish Proteins/chemistry , Zebrafish , Animals , Crystallography, X-Ray , Humans , Protein Domains
5.
Proc Natl Acad Sci U S A ; 110(34): 13916-20, 2013 08 20.
Article in English | MEDLINE | ID: mdl-23922389

ABSTRACT

Complement component C1, the complex that initiates the classical pathway of complement activation, is a 790-kDa assembly formed from the target-recognition subcomponent C1q and the modular proteases C1r and C1s. The proteases are elongated tetramers that become more compact when they bind to the collagen-like domains of C1q. Here, we describe a series of structures that reveal how the subcomponents associate to form C1. A complex between C1s and a collagen-like peptide containing the C1r/C1s-binding motif of C1q shows that the collagen binds to a shallow groove via a critical lysine side chain that contacts Ca(2+)-coordinating residues. The data explain the Ca(2+)-dependent binding mechanism, which is conserved in C1r and also in mannan-binding lectin-associated serine proteases, the serine proteases of the lectin pathway activation complexes. In an accompanying structure, C1s forms a compact ring-shaped tetramer featuring a unique head-to-tail interaction at its center that replicates the likely arrangement of C1r/C1s polypeptides in the C1 complex. Additional structures reveal how C1s polypeptides are positioned to enable activation by C1r and interaction with the substrate C4 inside the cage-like assembly formed by the collagenous stems of C1q. Together with previously determined structures of C1r fragments, the results reported here provide a structural basis for understanding the early steps of complement activation via the classical pathway.


Subject(s)
Complement Activation/immunology , Complement C1/chemistry , Complement C1q/chemistry , Complement C1s/chemistry , Immunity, Innate/immunology , Models, Molecular , Protein Conformation , Animals , CHO Cells , Chromatography, Affinity , Chromatography, Gel , Complement Activation/genetics , Complement C1q/metabolism , Complement C1s/metabolism , Cricetinae , Cricetulus , Crystallization , Escherichia coli , Protein Binding
6.
PLoS One ; 7(10): e47296, 2012.
Article in English | MEDLINE | ID: mdl-23077587

ABSTRACT

Oversulfated chondroitin sulfate (OSCS) has become the subject of multidisciplinary investigation as a non-traditional contaminant in the heparin therapeutic preparations that were linked to severe adverse events. In this study, it was found that OSCS inhibited complement fixation on bacteria and bacterial lysis mediated by the complement classical pathway. The inhibition of complement by OSCS is not due to interference with antibody/antigen interaction or due to consumption of C3 associated with FXII-dependent contact system activation. However, OSCS complement inhibition is dependent on C1 inhibitor (C1inh) since the depletion of C1inh from either normal or FXII-deficient complement plasma prevents OSCS inhibition of complement activity. Surface plasmon resonance measurements revealed that immobilized C1inhibitor bound greater than 5-fold more C1s in the presence of OSCS than in presence of heparin. Although heparin can also inhibit complement, OSCS and OSCS contaminated heparin are more potent inhibitors of complement. Furthermore, polysulfated glycosaminoglycan (PSGAG), an anti-inflammatory veterinary medicine with a similar structure to OSCS, also inhibited complement in the plasma of dogs and farm animals. This study provides a new insight that in addition to the FXII-dependent activation of contact system, oversulfated and polysulfated chondroitin-sulfate can inhibit complement activity by potentiating the classical complement pathway regulator C1inh. This effect on C1inh may play a role in inhibiting inflammation as well as impacting bacterial clearance.


Subject(s)
Chondroitin Sulfates/pharmacology , Complement C1 Inhibitor Protein/metabolism , Heparin/chemistry , Animals , Anticoagulants/adverse effects , Anticoagulants/chemistry , Blood Coagulation/drug effects , Chondroitin Sulfates/chemistry , Complement C1/antagonists & inhibitors , Complement C1/chemistry , Complement C1 Inhibitor Protein/chemistry , Dogs , Drug Contamination , Factor XII/chemistry , Factor XII/drug effects , Heparin/adverse effects , Plasma/chemistry , Plasma/metabolism
7.
ACS Nano ; 5(2): 730-7, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21214219

ABSTRACT

The classical pathway of complement is an essential component of the human innate immune system involved in the defense against pathogens as well as in the clearance of altered self-components. Activation of this pathway is triggered by C1, a multimolecular complex comprising a recognition protein C1q associated with a catalytic subunit C1s-C1r-C1r-C1s. We report here the direct observation of organized binding of C1 components C1q and C1s-C1r-C1r-C1s on carbon nanotubes, an ubiquitous component in nanotechnology research. Electron microscopy imaging showed individual multiwalled carbon nanotubes with protein molecules organized along the length of the sidewalls, often over 1 µm long. Less well-organized protein attachment was also observed on double-walled carbon nanotubes. Protein-solubilized nanotubes continued to attract protein molecules after their surface was fully covered. Despite the C1q binding properties, none of the nanotubes activated the C1 complex. We discuss these results on the adsorption mechanisms of macromolecules on carbon nanotubes and the possibility of using carbon nanotubes for structural studies of macromolecules. Importantly, the observations suggest that carbon nanotubes may interfere with the human immune system when entering the bloodstream. Our results raise caution in the applications of carbon nanotubes in biomedicine but may also open possibilities of novel applications concerning the many biochemical processes involving the versatile C1 macromolecule.


Subject(s)
Complement C1/chemistry , Immunity, Innate , Nanotubes, Carbon/chemistry , Complement C1/immunology , Complement C1/metabolism , Crystallization , Humans , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/immunology , Protein Subunits/metabolism , Surface Properties
8.
J Biol Chem ; 285(42): 32251-63, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20592021

ABSTRACT

C1, the complex that triggers the classic pathway of complement, is a 790-kDa assembly resulting from association of a recognition protein C1q with a Ca(2+)-dependent tetramer comprising two copies of the proteases C1r and C1s. Early structural investigations have shown that the extended C1s-C1r-C1r-C1s tetramer folds into a compact conformation in C1. Recent site-directed mutagenesis studies have identified the C1q-binding sites in C1r and C1s and led to a three-dimensional model of the C1 complex (Bally, I., Rossi, V., Lunardi, T., Thielens, N. M., Gaboriaud, C., and Arlaud, G. J. (2009) J. Biol. Chem. 284, 19340-19348). In this study, we have used a mass spectrometry-based strategy involving a label-free semi-quantitative analysis of protein samples to gain new structural insights into C1 assembly. Using a stable chemical modification, we have compared the accessibility of the lysine residues in the isolated tetramer and in C1. The labeling data account for 51 of the 73 lysine residues of C1r and C1s. They strongly support the hypothesis that both C1s CUB(1)-EGF-CUB(2) interaction domains, which are distant in the free tetramer, associate with each other in the C1 complex. This analysis also provides the first experimental evidence that, in the proenzyme form of C1, the C1s serine protease domain is partly positioned inside the C1q cone and yields precise information about its orientation in the complex. These results provide further structural insights into the architecture of the C1 complex, allowing significant improvement of our current C1 model.


Subject(s)
Complement C1/chemistry , Complement C1r/chemistry , Complement C1s/chemistry , Mass Spectrometry/methods , Protein Structure, Quaternary , Amino Acid Sequence , Binding Sites , Complement Activation , Complement C1/genetics , Complement C1/metabolism , Complement C1r/genetics , Complement C1r/metabolism , Complement C1s/genetics , Complement C1s/metabolism , Humans , Lysine/chemistry , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Binding , Staining and Labeling/methods , Surface Properties
9.
Folia Microbiol (Praha) ; 53(4): 295-302, 2008.
Article in English | MEDLINE | ID: mdl-18759112

ABSTRACT

DNA aptamers were developed against lipopolysaccharide (LPS) from E. coli O111:B4 and shown to bind both LPS and E. coli by a colorimetric enzyme-based microplate assay. The polyclonal aptamers were coupled to human C1qrs protein either directly using a bifunctional linker or indirectly using biotinylated aptamers and a streptavidin-C1qrs complex. Both systems significantly reduced colony counts when applied to E. coli O111:B4 and K12 strains across a series of 10x dilutions of the bacteria in the presence of human serum; it was diluted 1: 10(3) in order to avoid significant bacterial lysis by the competing alternate pathway of complement activation. A number of candidate DNA aptamer sequences were cloned and sequenced from the anti-LPS aptamer library for future screening of antibacterial or "antibiotic" potential and to aid in eventual development of an alternative therapy for antibiotic-resistant bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Aptamers, Nucleotide/pharmacology , Complement C1/immunology , Escherichia coli/immunology , Lipopolysaccharides/immunology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/immunology , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/immunology , Base Sequence , Complement C1/chemistry , Escherichia coli/drug effects , Escherichia coli Infections/drug therapy , Escherichia coli Infections/immunology , Humans , Lipopolysaccharides/chemistry , Molecular Sequence Data , SELEX Aptamer Technique
10.
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
11.
Immunobiology ; 212(4-5): 279-88, 2007.
Article in English | MEDLINE | ID: mdl-17544813

ABSTRACT

The classical pathway C1 complex, and the MBL-MASP and ficolin-MASP complexes involved in activation of the lectin pathway have several features in common. Both types of complexes are assembled from two subunits: an oligomeric recognition protein (C1q, MBL, L-, H- or M-ficolin), and a protease component, which is either a tetramer (C1s-C1r-C1r-C1s) or a dimer ((MASP)(2)). Recent functional and 3-D structural investigations have revealed that C1r/C1s and the MASPs associate through a common mechanism involving their N-terminal CUB1-EGF region. In contrast, the C1s-C1r-C1r-C1s tetramer and the (MASP)(2) dimers appear to have evolved distinct strategies to associate with their partner proteins. The purpose of this article is to review these recent advances.


Subject(s)
Complement C1/metabolism , Lectins/metabolism , Mannose-Binding Lectin/metabolism , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Animals , Complement C1/chemistry , Humans , Lectins/chemistry , Mannose-Binding Lectin/chemistry , Mannose-Binding Protein-Associated Serine Proteases/chemistry , Models, Molecular , Protein Binding , Ficolins
12.
Mol Immunol ; 43(7): 800-11, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16098595

ABSTRACT

The fourth component of human complement is an essential part of the classical and lectin pathways performing multifunctional roles in both host defense and immune regulation. C4 is the most polymorphic member of the complement proteins, and complete deficiency is strongly associated with autoimmune disease, especially, systemic lupus erythematosus (SLE). Of the two C4 genes C4A, but not C4B, null alleles have been implicated as important independent disease susceptibility genes occurring in more than half of SLE patients. Whether and how this deficiency contributes to the development or pathology remains unclear. We do know that activation of C4 by C1s cleaves the thioester bond, thus inducing a conformational change that exposes numerous ligand-binding sites involved in functional activity. Structural comparison, among many other tools, plays an important role in predicting function. In this report, the tertiary structures of C4A and C4B were compared using near and far-UV circular dichroism, ANS fluorescence, site-specific monoclonal antibodies and isoelectric focusing. Negligible differences in the native proteins were found. However, the activated proteins were dissimilar in secondary and tertiary structure that was accompanied by significant differences in charge distribution and surface hydrophobicity. These conformational differences, together with known acceptor preferences, have functional implications for the association between C4A null alleles and SLE.


Subject(s)
Complement Activation , Complement C4a/chemistry , Complement C4b/chemistry , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Alleles , Antibodies, Monoclonal , Circular Dichroism , Complement C1/chemistry , Complement C4a/genetics , Complement C4a/immunology , Complement C4b/genetics , Complement C4b/immunology , Fluorescence , Humans , Isoelectric Focusing , Protein Structure, Tertiary , Spectroscopy, Near-Infrared
14.
Thromb Res ; 113(3-4): 243-50, 2004.
Article in English | MEDLINE | ID: mdl-15140589

ABSTRACT

Dermatan sulfate (DS) is a member of the family of structurally complex, sulfated, linear heteropolysaccharides called glycosaminoglycans (GAGs). It has a similar structure to heparin and heparan sulfate (HS), but with acetylgalactosamine replacing glucosamine, and the uronic acid moiety, mainly iduronic, joined 1-->3 to the hexosamine. We are studying the relationships between structure and activities of dermatan sulfate, in particular those associated with the thrombin inhibition mediated by heparin cofactor II (HCII). As we have demonstrated with heparin, a small fraction of dermatan sulfate was isolated by precipitation with the first component of the complement system, under very specific conditions of low ionic strength, and the presence of calcium ions. The sulfate content and the anticoagulant activity of the dermatan sulfate fraction isolated in the precipitate were three and four times greater respectively than the starting material. Our in vivo studies showed that this fraction has threefold higher thrombolytic activity than the DS. All these results suggest that this fraction could be used as a therapeutic agent for thrombi dissolution.


Subject(s)
Anticoagulants/chemistry , Anticoagulants/pharmacology , Complement C1/metabolism , Dermatan Sulfate/chemistry , Dermatan Sulfate/pharmacology , Acetylgalactosamine/chemistry , Animals , Anticoagulants/isolation & purification , Anticoagulants/metabolism , Calcium/chemistry , Chemical Precipitation , Complement C1/chemistry , Complement C1/isolation & purification , Dermatan Sulfate/isolation & purification , Dermatan Sulfate/metabolism , Fibrinolytic Agents/pharmacology , Hexosamines/chemistry , Iduronic Acid/chemistry , Male , Osmolar Concentration , Rats , Rats, Wistar , Structure-Activity Relationship , Sulfates/chemistry
15.
Biochemistry ; 42(50): 14939-45, 2003 Dec 23.
Article in English | MEDLINE | ID: mdl-14674770

ABSTRACT

The classical complement pathway, which plays a vital role in preventing infection, is initiated by the action of the serine proteases C1r and C1s. We have examined the hydrolysis of substrates representing cleavage sequences in the physiological substrates for C1s, C2 and C4. These studies showed that the P(1)'-P(4)' substrate residues of C2 and C4 conferred greater affinity of substrate for enzyme and also induced a sigmoidal dependence of enzyme velocity on substrate concentration. This indicates that the substrate gave rise to homotropic positive cooperative behavior in the enzyme. When C1s was in complex with C1q and C1r, as would occur under physiological conditions, the same behavior was observed, indicating that this mechanism is relevant in the complement pathway in vivo. We further investigated the requirements of C1s for prime side amino acids by examining a substrate library in which each of the P(1)'-P(4)' positions had been substituted by different classes of amino acids. This revealed that the P(1)' position was a major determinant of the selectivity of the enzyme, while certain substitutions at the P(1)'-P(4)' positions abolished the allosteric behavior, indicating that contact residues at these positions in the C1s enzyme must mediate the cooperativity. The studies reported here highlight the importance of prime subsites in C1s for interaction with its cognate substrates in the complement pathway and therefore yield greater understanding of the mechanism of interaction between this vital protease and its physiological substrates.


Subject(s)
Complement C1s/chemistry , Complement Pathway, Classical , Serine Endopeptidases/chemistry , Amino Acid Substitution , Binding Sites , Complement C1/chemistry , Complement C1/metabolism , Complement C1s/metabolism , Complement C2/chemistry , Complement C2/metabolism , Complement C4/chemistry , Complement C4/metabolism , Coumarins/chemical synthesis , Coumarins/metabolism , Humans , Hydrolysis , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Peptide Library , Serine Endopeptidases/metabolism , Substrate Specificity
16.
J Biol Chem ; 278(34): 32157-64, 2003 Aug 22.
Article in English | MEDLINE | ID: mdl-12788922

ABSTRACT

C1, the complex that triggers the classical pathway of complement, is assembled from two modular proteases C1r and C1s and a recognition protein C1q. The N-terminal CUB1-EGF segments of C1r and C1s are key elements of the C1 architecture, because they mediate both Ca2+-dependent C1r-C1s association and interaction with C1q. The crystal structure of the interaction domain of C1s has been solved and refined to 1.5 A resolution. The structure reveals a head-to-tail homodimer involving interactions between the CUB1 module of one monomer and the epidermal growth factor (EGF) module of its counterpart. A Ca2+ ion is bound to each EGF module and stabilizes both the intra- and inter-monomer interfaces. Unexpectedly, a second Ca2+ ion is bound to the distal end of each CUB1 module, through six ligands contributed by Glu45, Asp53, Asp98, and two water molecules. These acidic residues and Tyr17 are conserved in approximately two-thirds of the CUB repertoire and define a novel, Ca2+-binding CUB module subset. The C1s structure was used to build a model of the C1r-C1s CUB1-EGF heterodimer, which in C1 connects C1r to C1s and mediates interaction with C1q. A structural model of the C1q/C1r/C1s interface is proposed, where the rod-like collagen triple helix of C1q is accommodated into a groove along the transversal axis of the C1r-C1s heterodimer.


Subject(s)
Calcium/metabolism , Complement C1/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Complement C1/chemistry , Crystallography, X-Ray , DNA Primers , Dimerization , Electrophoresis, Polyacrylamide Gel , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Spodoptera
17.
Mol Immunol ; 39(7-8): 383-94, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12413689

ABSTRACT

C1 is the multimolecular protease that triggers activation of the classical pathway of complement, a major element of antimicrobial host defense also involved in immune tolerance and various pathologies. This 790,000 Da complex is formed from the association of a recognition protein, C1q, and a catalytic subunit, the Ca2+-dependent tetramer C1s-C1r-C1r-C1s comprising two copies of each of the modular proteases C1r and C1s. Early studies mainly based on biochemical analysis and electron microscopy of C1 and its isolated components have allowed for characterization of their domain structure and led to a low-resolution model of the C1 complex in which the elongated C1s-C1r-C1r-C1s tetramer folds into a more compact, "8-shaped" conformation upon interaction with C1q. A major strategy used over the past years has been to dissect the C1 proteins into modular segments to characterize their function and solve their structure by either X-ray crystallography or nuclear magnetic resonance spectroscopy (NMR). The purpose of this review is to focus on this information, with particular emphasis on the architecture of the C1 complex and the mechanisms underlying its activation and proteolytic activity.


Subject(s)
Complement Activation , Complement C1/chemistry , Animals , Catalytic Domain , Complement C1/physiology , Complement C1q/chemistry , Crystallography, X-Ray , Epidermal Growth Factor/chemistry , Humans , Magnetic Resonance Spectroscopy , Protein Structure, Secondary , Serine Endopeptidases/chemistry
18.
Biochem Soc Trans ; 30(Pt 6): 1001-6, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12440961

ABSTRACT

The classical complement pathway is a major element of innate immunity against infection, and is also involved in immune tolerance, graft rejection and various pathologies. This pathway is triggered by C1, a multimolecular protease formed from the association of a recognition protein, C1q, and a catalytic subunit, the calcium-dependent tetramer C1s-C1r-C1r-C1s, which comprises two copies of each of the modular proteases C1r and C1s. All activators of the pathway are recognized by the C1q moiety of C1, a process that generates a conformational signal that triggers self-activation of C1r, which in turn activates C1s, the enzyme that mediates specific cleavage of C4 and C2, the C1 substrates. Early work based on biochemical and electron microscopy studies has allowed characterization of the domain structure of the C1 subcomponents and led to a low-resolution model of the complex in which the elongated C1s-C1r-C1r-C1s tetramer folds into a compact, figure-of-8-shaped conformation upon interaction with C1q. The strategy used over the past decade was based on a dissection of the C1 proteins into modular segments to characterize their function and solve their three-dimensional structure by X-ray crystallography or NMR spectroscopy. This approach allows deep insights into the structure-function relationships of C1, particularly with respect to the assembly of the C1 complex and the mechanisms underlying its activation and proteolytic activity.


Subject(s)
Complement C1/chemistry , Complement C1/physiology , Animals , Catalytic Domain , Complement C1/metabolism , Enzyme Precursors/chemistry , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary
20.
EMBO J ; 21(3): 231-9, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11823416

ABSTRACT

C1r is the modular serine protease (SP) that mediates autolytic activation of C1, the macromolecular complex that triggers the classical pathway of complement. The crystal structure of a mutated, proenzyme form of the catalytic domain of human C1r, comprising the first and second complement control protein modules (CCP1, CCP2) and the SP domain has been solved and refined to 2.9 A resolution. The domain associates as a homodimer with an elongated head-to-tail structure featuring a central opening and involving interactions between the CCP1 module of one monomer and the SP domain of its counterpart. Consequently, the catalytic site of one monomer and the cleavage site of the other are located at opposite ends of the dimer. The structure reveals unusual features in the SP domain and provides strong support for the hypothesis that C1r activation in C1 is triggered by a mechanical stress caused by target recognition that disrupts the CCP1-SP interfaces and allows formation of transient states involving important conformational changes.


Subject(s)
Complement C1/metabolism , Complement C1r/chemistry , Amino Acid Sequence , Catalysis , Complement C1/chemistry , Complement C1r/genetics , Complement C1r/metabolism , Complement Pathway, Classical , Enzyme Precursors/chemistry , Enzyme Precursors/genetics , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Alignment , Stress, Mechanical , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...