Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Front Immunol ; 12: 714055, 2021.
Article in English | MEDLINE | ID: mdl-34434196

ABSTRACT

Cleavage of C3 to C3a and C3b plays a central role in the generation of complement-mediated defences. Although the thioester-mediated surface deposition of C3b has been well-studied, fluid phase dimers of C3 fragments remain largely unexplored. Here we show C3 cleavage results in the spontaneous formation of C3b dimers and present the first X-ray crystal structure of a disulphide-linked human C3d dimer. Binding studies reveal these dimers are capable of crosslinking complement receptor 2 and preliminary cell-based analyses suggest they could modulate B cell activation to influence tolerogenic pathways. Altogether, insights into the physiologically-relevant functions of C3d(g) dimers gained from our findings will pave the way to enhancing our understanding surrounding the importance of complement in the fluid phase and could inform the design of novel therapies for immune system disorders in the future.


Subject(s)
Complement C3d/chemistry , Models, Molecular , Protein Multimerization , Complement C3/chemistry , Complement C3/immunology , Complement C3d/immunology , Humans , Lymphocyte Activation/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Conformation , Proteolysis , Recombinant Proteins/chemistry , Structure-Activity Relationship
2.
J Biol Chem ; 294(52): 20148-20163, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31719147

ABSTRACT

Activation and suppression of the complement system compete on every serum-exposed surface, host or foreign. Potentially harmful outcomes of this competition depend on surface molecules through mechanisms that remain incompletely understood. Combining surface plasmon resonance (SPR) with atomic force microscopy (AFM), here we studied two complement system proteins at the single-molecule level: C3b, the proteolytically activated form of C3, and factor H (FH), the surface-sensing C3b-binding complement regulator. We used SPR to monitor complement initiation occurring through a positive-feedback loop wherein surface-deposited C3b participates in convertases that cleave C3, thereby depositing more C3b. Over multiple cycles of flowing factor B, factor D, and C3 over the SPR chip, we amplified C3b from ∼20 to ∼220 molecules·µm-2 AFM revealed C3b clusters of up to 20 molecules and solitary C3b molecules deposited up to 200 nm away from the clusters. A force of 0.17 ± 0.02 nanonewtons was needed to pull a single FH molecule, anchored to the AFM probe, from its complex with surface-attached C3b. The extent to which FH molecules stretched before detachment varied widely among complexes. Performing force-distance measurements with FH(D1119G), a variant lacking one of the C3b-binding sites and causing atypical hemolytic uremic syndrome, we found that it detached more uniformly and easily. In further SPR experiments, KD values between FH and C3b on a custom-made chip surface were 5-fold tighter than on commercial chips and similar to those on erythrocytes. These results suggest that the chemistry at the surface on which FH acts drives conformational adjustments that are functionally critical.


Subject(s)
Complement C3b/metabolism , Complement Factor H/metabolism , Microscopy, Atomic Force , Surface Plasmon Resonance , Complement Activation , Complement C3b/chemistry , Complement C3d/chemistry , Complement C3d/metabolism , Complement Factor H/chemistry , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Kinetics , Protein Binding
3.
J Struct Biol ; 208(2): 77-85, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31400508

ABSTRACT

The gram-negative bacterium Moraxella catarrhalis infects humans exclusively, causing various respiratory tract diseases, including acute otitis media in children, septicaemia or meningitis in adults, and pneumonia in the elderly. To do so, M. catarrhalis expresses virulence factors facilitating its entry and survival in the host. Among them are the ubiquitous surface proteins (Usps): A1, A2, and A2H, which all belong to the trimeric autotransporter adhesin family. They bind extracellular matrix molecules and inhibit the classical and alternative pathways of the complement cascade by recruiting complement regulators C3d and C4b binding protein. Here, we report the 2.5 Šresolution X-ray structure of UspA1299-452, which previous work had suggested contained the canonical C3d binding site found in both UspA1 and UspA2. We show that this fragment of the passenger domain contains part of the long neck domain (residues 299-336) and a fragment of the stalk (residues 337-452). The coiled-coil stalk is left-handed, with 7 polar residues from each chain facing the core and coordinating chloride ions or water molecules. Despite the previous reports of tight binding in serum-based assays, we were not able to demonstrate binding between C3d and UspA1299-452 using ELISA or biolayer interferometry, and the two proteins run separately on size-exclusion chromatography. Microscale thermophoresis suggested that the dissociation constant was 140.5 ±â€¯8.4 µM. We therefore suggest that full-length proteins or other additional factors are important in UspA1-C3d interactions. Other molecules on the bacterial surface or present in serum may enhance binding of those two molecules.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Complement C3d/chemistry , Complement C3d/metabolism , Moraxella catarrhalis/metabolism , Anisotropy , Binding Sites , Chromatography, Gel , Crystallography, X-Ray , Protein Binding , Protein Structure, Secondary
4.
Chem Biol Drug Des ; 93(4): 628-637, 2019 04.
Article in English | MEDLINE | ID: mdl-30566277

ABSTRACT

The interactions of complement receptor 2 (CR2) and the degradation fragment C3d of complement component C3 mediate the innate and adaptive immune systems. Due to the importance of C3d-CR2 interaction in the design of vaccines, many studies have indicated the interactions are pH-dependent. Moreover, C3d-CR2 interactions at pH 5.0 are unknown. To investigate the molecular mechanism of pH-regulating C3d-CR2 interaction, molecular dynamics simulations for C3d-CR2 complex in different pH are performed. Our results revealed that the protonation of His9 in C3d at pH 6.0 slightly weakens C3d-CR2 association as reducing pH from 7.4 to 6.0, initiated from a key hydrogen bond formed between Gly270 and His9 in C3d at pH 6.0. When reducing pH from 6.0 to 5.0, the protonation of His33 in C3d weakens C3d-SCR1 association by changing the hydrogen-bond network of Asp36, Glu37, and Glu39 in C3d with Arg13 in CR2. In addition, the protonation of His90 significantly enhances C3d-SCR2 association. This is because the enhanced hydrogen-bond interactions of His90 with Glu63 and Ser69 of the linker change the conformations of the linker, Cys112-Asn116 and Pro87-Gly91 regions. This study uncovers the molecular mechanism of the mediation of pH on C3d-CR2 interaction, which is valuable for vaccine design.


Subject(s)
Complement C3d/metabolism , Molecular Dynamics Simulation , Receptors, Complement 3d/metabolism , Binding Sites , Complement C3d/chemistry , Humans , Hydrogen-Ion Concentration , Protein Binding , Protein Structure, Tertiary , Receptors, Complement 3d/chemistry , Thermodynamics
5.
Transpl Int ; 31(11): 1189-1197, 2018 11.
Article in English | MEDLINE | ID: mdl-30091231

ABSTRACT

Luminex single antigen bead assays revolutionized human leukocyte antigen (HLA) antibody detection owing to their superior sensitivity compared to conventional methods. Nevertheless, the advent of higher sensitivity came at the expense of difficulty in clinical decision-making, since not all luminex detectable antibodies are clinically relevant. Therefore, new tools such as C1q/C3d assays and IgG subclass analysis emerged with the aim to discriminate the inert antibodies from the deleterious ones. Here, we provide an overview on the technical challenges related to these different HLA antibody detection systems and briefly refer to the recent literature regarding the clinical relevance of these assays, mainly in the field of kidney transplantation.


Subject(s)
Complement C1q/chemistry , Complement C3d/chemistry , HLA Antigens/chemistry , Histocompatibility Testing/methods , Immunoglobulin G/chemistry , Decision Support Systems, Clinical , Graft Rejection/immunology , Humans , Isoantibodies/immunology , Kidney Transplantation , Postoperative Period , Prognosis , Protein Binding , Risk
6.
J Biol Chem ; 292(32): 13345-13360, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28637873

ABSTRACT

Spontaneous activation enables the complement system to respond very rapidly to diverse threats. This activation is efficiently suppressed by complement factor H (CFH) on self-surfaces but not on foreign surfaces. The surface selectivity of CFH, a soluble protein containing 20 complement-control protein modules (CCPs 1-20), may be compromised by disease-linked mutations. However, which of the several functions of CFH drives this self-surface selectivity remains unknown. To address this, we expressed human CFH mutants in Pichia pastoris We found that recombinant I62-CFH (protective against age-related macular degeneration) and V62-CFH functioned equivalently, matching or outperforming plasma-derived CFH, whereas R53H-CFH, linked to atypical hemolytic uremic syndrome (aHUS), was defective in C3bBb decay-accelerating activity (DAA) and factor I cofactor activity (CA). The aHUS-linked CCP 19 mutant D1119G-CFH had virtually no CA on (self-like) sheep erythrocytes (ES) but retained DAA. The aHUS-linked CCP 20 mutant S1191L/V1197A-CFH (LA-CFH) had dramatically reduced CA on ES but was less compromised in DAA. D1119G-CFH and LA-CFH both performed poorly at preventing complement-mediated hemolysis of ES PspCN, a CFH-binding Streptococcus pneumoniae protein domain, binds CFH tightly and increases accessibility of CCPs 19 and 20. PspCN did not improve the DAA of any CFH variant on ES Conversely, PspCN boosted the CA, on ES, of I62-CFH, R53H-CFH, and LA-CFH and also enhanced hemolysis protection by I62-CFH and LA-CFH. We conclude that CCPs 19 and 20 are critical for efficient CA on self-surfaces but less important for DAA. Exposing CCPs 19 and 20 with PspCN and thus enhancing CA on self-surfaces may reverse deficiencies of some CFH variants.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Complement Activation , Macular Degeneration/genetics , Mutation , Amino Acid Substitution , Animals , Atypical Hemolytic Uremic Syndrome/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Complement C3 Convertase, Alternative Pathway/chemistry , Complement C3 Convertase, Alternative Pathway/genetics , Complement C3 Convertase, Alternative Pathway/metabolism , Complement C3d/chemistry , Complement C3d/genetics , Complement C3d/metabolism , Complement Factor H/chemistry , Complement Factor H/genetics , Complement Factor H/metabolism , Complement Factor I/chemistry , Complement Factor I/genetics , Complement Factor I/metabolism , Erythrocytes/chemistry , Hemolysis , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/genetics , Immobilized Proteins/metabolism , Macular Degeneration/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sheep, Domestic , Solubility , Streptococcus pneumoniae/metabolism , Surface Properties
7.
J Phys Chem B ; 120(33): 8416-23, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27092816

ABSTRACT

Electrostatic effects are ubiquitous in protein interactions and are found to be pervasive in the complement system as well. The interaction between complement fragment C3d and complement receptor 2 (CR2) has evolved to become a link between innate and adaptive immunity. Electrostatic interactions have been suggested to be the driving factor for the association of the C3d:CR2 complex. In this study, we investigate the effects of ionic strength and mutagenesis on the association of C3d:CR2 through Brownian dynamics simulations. We demonstrate that the formation of the C3d:CR2 complex is ionic strength-dependent, suggesting the presence of long-range electrostatic steering that accelerates the complex formation. Electrostatic steering occurs through the interaction of an acidic surface patch in C3d and the positively charged CR2 and is supported by the effects of mutations within the acidic patch of C3d that slow or diminish association. Our data are in agreement with previous experimental mutagenesis and binding studies and computational studies. Although the C3d acidic patch may be locally destabilizing because of unfavorable Coulombic interactions of like charges, it contributes to the acceleration of association. Therefore, acceleration of function through electrostatic steering takes precedence to stability. The site of interaction between C3d and CR2 has been the target for delivery of CR2-bound nanoparticle, antibody, and small molecule biomarkers, as well as potential therapeutics. A detailed knowledge of the physicochemical basis of C3d:CR2 association may be necessary to accelerate biomarker and drug discovery efforts.


Subject(s)
Complement C3d/metabolism , Receptors, Complement 3d/metabolism , Complement C3d/chemistry , Complement C3d/genetics , Computer Simulation , Models, Molecular , Mutation , Protein Binding , Receptors, Complement 3d/chemistry , Receptors, Complement 3d/genetics , Static Electricity
8.
Protein Sci ; 24(5): 789-802, 2015 May.
Article in English | MEDLINE | ID: mdl-25628052

ABSTRACT

As a part of innate immunity, the complement system relies on activation of the alternative pathway (AP). While feed-forward amplification generates an immune response towards foreign surfaces, the process requires regulation to prevent an immune response on the surface of host cells. Factor H (FH) is a complement protein secreted by native cells to negatively regulate the AP. In terms of structure, FH is composed of 20 complement-control protein (CCP) modules that are structurally homologous but vary in composition and function. Mutations in these CCPs have been linked to states of autoimmunity. In particular, several mutations in CCP 19-20 are correlated to atypical hemolytic uremic syndrome (aHUS). From crystallographic structures there are three putative binding sites of CCP 19-20 on C3d. Since there has been some controversy over the primary mode of binding from experimental studies, we approach characterization of binding using computational methods. Specifically, we compare each binding mode in terms of electrostatic character, structural stability, dissociative and associative properties, and predicted free energy of binding. After a detailed investigation, we found two of the three binding sites to be similarly stable while varying in the number of contacts to C3d and in the energetic barrier to complex dissociation. These sites are likely physiologically relevant and may facilitate multivalent binding of FH CCP 19-20 to C3b and either C3d or host glycosaminoglycans. We propose thermodynamically stable binding with modules 19 and 20, the latter driven by electrostatics, acting synergistically to increase the apparent affinity of FH for host surfaces.


Subject(s)
Complement C3d/chemistry , Complement Factor H/chemistry , Immunity, Innate , Protein Structure, Tertiary , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/immunology , Binding Sites , Complement C3 Convertase, Alternative Pathway/chemistry , Complement C3d/genetics , Complement C3d/immunology , Complement Factor H/genetics , Complement Factor H/immunology , Humans , Models, Molecular , Mutation , Protein Binding , Structural Homology, Protein
9.
Mol Immunol ; 64(1): 112-22, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25433434

ABSTRACT

The C3d:CR2(SCR1-2) interaction plays an important role in bridging innate and adaptive immunity, leading to enhanced antibody production at sites of complement activation. Over the past decade, there has been much debate over the binding mode of this interaction. An initial cocrystal structure (PDB: 1GHQ) was published in 2001, in which the only interactions observed were between the SCR2 domain of CR2 and a side-face of C3d whereas a cocrystal structure (PDB: 3OED) published in 2011 showed both the SCR1 and SCR2 domains of CR2 interacting with an acidic patch on the concave surface of C3d. The initial 1GHQ structure is at odds with the majority of existing biochemical data and the publication of the 3OED structure renewed uncertainty regarding the physiological relevance of 1GHQ, suggesting that crystallization may have been influenced by the presence of zinc acetate in the crystallization process. In our study, we used a variety of computational approaches to gain insight into the binding mode between C3d and CR2 and demonstrate that the binding site at the acidic patch (3OED) is electrostatically more favorable, exhibits better structural and dissociative stability, specifically at the SCR1 domain, and has higher binding affinity than the 1GHQ binding mode. We also observe that nonphysiological zinc ions enhance the formation of the C3d:CR2 complex at the side face of C3d (1GHQ) through increases in electrostatic favorability, intermolecular interactions, dissociative character and overall energetic favorability. These results provide a theoretical basis for the association of C3d:CR2 at the acidic cavity of C3d and provide an explanation for binding of CR2 at the side face of C3d in the presence of nonphysiological zinc ions.


Subject(s)
Complement C3d/metabolism , Models, Immunological , Models, Molecular , Receptors, Complement 3d/metabolism , Complement C3d/chemistry , Molecular Dynamics Simulation , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Receptors, Complement 3d/chemistry , Solvents/chemistry , Static Electricity
10.
Transplantation ; 98(6): 625-31, 2014 Sep 27.
Article in English | MEDLINE | ID: mdl-25058376

ABSTRACT

BACKGROUND: Anti-human leukocyte antigen (HLA) antibody detection in solid-phase flow beads assays can be quenched by complement activation, but the precise mechanism of this interference is not fully elucidated yet. METHODS: Using the Luminex flow beads screening assay for detection of anti-HLA antibodies, we analyzed the binding of high concentrations of the pan class I anti-HLA monoclonal antibody W6/32 in neat normal, ethylenediaminetetraacetic acid-treated normal and complement factors C1q, C4/C3, C2, C3, factor B or C5-depleted human sera, using anti-mouse immunoglobulin G as the detection antibody. Complement activation and binding to beads were revealed using anti-human C1q, C4d, and C3d antibodies. To translate our findings to the human setting, we used the class I and class II HLA single-antigen flow beads assays and sera from four patients with high titers of antibodies. RESULTS: Detection of W6/32 did not suffer any interference with C1q and C4/C3-depleted sera. A partial quenching was observed with C2, C3, and factor B-depleted sera, but was more pronounced with the factor B-depleted serum. W6/32 was undetectable in presence of C5-depleted serum. The binding of activation products derived from C3 principally, and also from C4, impaired immunoglobulin G and C1q detection. Accordingly, C4d detection was hindered by deposition of activated C3. Similar findings were obtained with patients' sera. CONCLUSION: Binding of C4 and C3 activation products is the main responsible for complement interference in flow beads assays. A complete quenching requires complement activation through C3 cleavage and its amplification by the alternative pathway.


Subject(s)
Complement System Proteins/chemistry , HLA Antigens/chemistry , HLA Antigens/immunology , Immunoassay/methods , Antibodies, Monoclonal/chemistry , Complement Activation , Complement C1q/chemistry , Complement C3/deficiency , Complement C3d/chemistry , Complement C4b/chemistry , Complement System Proteins/immunology , Edetic Acid/chemistry , Hereditary Complement Deficiency Diseases , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunologic Deficiency Syndromes , Peptide Fragments/chemistry , Protein Binding
11.
Biophys J ; 106(5): 1164-73, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24606940

ABSTRACT

Staphylococcus aureus expresses numerous virulence factors that aid in immune evasion. The four-domain staphylococcal immunoglobulin binding (Sbi) protein interacts with complement component 3 (C3) and its thioester domain (C3d)-containing fragments. Recent structural data suggested two possible modes of binding of Sbi domain IV (Sbi-IV) to C3d, but the physiological binding mode remains unclear. We used a computational approach to provide insight into the C3d-Sbi-IV interaction. Molecular dynamics (MD) simulations showed that the first binding mode (PDB code 2WY8) is more robust than the second (PDB code 2WY7), with more persistent polar and nonpolar interactions, as well as conserved interfacial solvent accessible surface area. Brownian dynamics and steered MD simulations revealed that the first binding mode has faster association kinetics and maintains more stable intermolecular interactions compared to the second binding mode. In light of available experimental and structural data, our data confirm that the first binding mode represents Sbi-IV interaction with C3d (and C3) in a physiological context. Although the second binding mode is inherently less stable, we suggest a possible physiological role. Both binding sites may serve as a template for structure-based design of novel complement therapeutics.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Complement C3d/metabolism , Molecular Dynamics Simulation , Virulence Factors/metabolism , Bacterial Proteins/chemistry , Carrier Proteins/chemistry , Complement C3d/chemistry , Crystallography, X-Ray , Protein Binding , Protein Conformation , Protein Stability , Static Electricity , Virulence Factors/chemistry
12.
Phys Chem Chem Phys ; 15(4): 1241-51, 2013 Jan 28.
Article in English | MEDLINE | ID: mdl-23229122

ABSTRACT

The interaction between human complement receptor type 2 (CR2) and antigen-bound C3d can bridge the innate and adaptive immune systems. The recently determined structure of the CR2(SCR1-2):C3d complex has revealed the expected binding interface of CR2-C3d. In this article, wild type (WT) and three mutants of the new structure are studied by molecular dynamics (MD) simulations. The differently decreased structural stabilities of the mutants relative to WT are shown to be consistent with the experimental data, which can be explained by the different hydrogen bond patterns at the interfaces. It is also found that two clusters of residues (D36/E37/E39 and E160/D163/E166) in the acidic pocket of C3d are important for CR2-C3d interactions, which is in good agreement with previous mutagenesis study. In addition, functional dynamics and the conformational change of CR2 are explored by using domain cross-correlation map (DCCM), principal component analysis (PCA), and free energy landscape (FEL) methods. The conformational change mainly corresponds to the opening of a V-shaped structure of CR2, which is consistent with the previously reported high interdomain flexibility of CR2. We further suppose that the opening of a V-shaped structure of CR2 may favor the binding stability of CR2(SCR1-2):C3d. This study would provide some new insights into the understanding of the CR2-C3d interaction mechanism.


Subject(s)
Complement C3d/metabolism , Molecular Dynamics Simulation , Receptors, Complement 3d/metabolism , Complement C3d/chemistry , Complement C3d/genetics , Humans , Mutagenesis , Principal Component Analysis , Protein Binding , Protein Structure, Tertiary , Receptors, Complement 3d/chemistry , Receptors, Complement 3d/genetics , Thermodynamics
13.
Immunity ; 37(2): 199-207, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22921118

ABSTRACT

The complement system of innate immunity is important in regulating humoral immunity largely through the complement receptor CR2, which forms a coreceptor on B cells during antigen-induced activation. However, CR2 also retains antigens on follicular dendritic cells (FDCs). Display of antigen on FDCs is critical for clonal selection and affinity maturation of activated B cells. This review will discuss the role of complement in adaptive immunity in general with a focus on the interplay between CR2-associated antigen on B cells with CR2 expressed on FDCs. This latter interaction provides an opportunity for memory B cells to sample antigen over prolonged periods. The cocrystal structure of CR2 with its ligand C3d provides insight into how the complement system regulates access of antigen by B cells with implications for therapeutic manipulations to modulate aberrant B cell responses in the case of autoimmunity.


Subject(s)
B-Lymphocytes/immunology , Complement C3d/immunology , Dendritic Cells, Follicular/immunology , Immunity, Humoral/immunology , Receptors, Complement 3d/immunology , Animals , Antigen Presentation/immunology , Antigens/immunology , Antigens, CD19/immunology , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Complement Activation/immunology , Complement C3d/chemistry , Complement C3d/metabolism , Dendritic Cells, Follicular/metabolism , Humans , Immunity, Innate , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Receptors, Complement 3d/chemistry , Receptors, Complement 3d/metabolism , Tetraspanin 28/immunology
14.
J Mol Biol ; 418(3-4): 248-63, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22387467

ABSTRACT

A novel therapeutic reagent TT30 was designed to be effective in diseases of the alternative pathway of complement such as paroxysmal nocturnal hemoglobinuria and other diseases. TT30 is constructed from the first four short complement regulator (SCR) domains of complement receptor type 2 (CR2) that bind to complement C3d, followed by the first five SCR domains of complement factor H that bind to complement C3b. In order to assess how TT30 binds to C3d and C3b, we determined the TT30 solution structure by a combination of analytical ultracentrifugation, X-ray scattering and constrained modeling. The sedimentation coefficients and radius of gyration of TT30 were unaffected by citrate or phosphate-buffered saline buffers and indicate an elongated monomeric structure with a sedimentation coefficient of 3.1 S and a radius of gyration R(G) of 6.9 nm. Molecular modeling starting from 3000 randomized TT30 conformations showed that high-quality X-ray curve fits were obtained with extended SCR arrangements, showing that TT30 has a limited degree of inter-SCR flexibility in its solution structure. The best-fit TT30 structural models are readily merged with the crystal structure of C3b to show that the four CR2 domains extend freely into solution when the five complement factor H domains are bound within C3b. We reevaluated the solution structure of the CR2-C3d complex that confirmed its recent crystal structure. This recent CR2-C3d crystal structure showed that TT30 is able to interact readily with C3d ligands in many orientations when TT30 is bound to C3b.


Subject(s)
Complement C3b/chemistry , Complement C3d/chemistry , Recombinant Proteins/chemistry , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Complement C3b/metabolism , Complement C3d/metabolism , Cricetinae , Crystallography, X-Ray , Ligands , Models, Molecular , Molecular Sequence Data , Protein Conformation , Receptors, Complement 3d/chemistry , Receptors, Complement 3d/genetics , Receptors, Complement 3d/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Solutions , Ultracentrifugation
15.
Biochemistry ; 51(9): 1874-84, 2012 Mar 06.
Article in English | MEDLINE | ID: mdl-22320225

ABSTRACT

Numerous complement factor H (FH) mutations predispose patients to atypical hemolytic uremic syndrome (aHUS) and other disorders arising from inadequately regulated complement activation. No unifying structural or mechanistic consequences have been ascribed to these mutants beyond impaired self-cell protection. The S1191L and V1197A mutations toward the C-terminus of FH, which occur in patients singly or together, arose from gene conversion between CFH encoding FH and CFHR1 encoding FH-related 1. We show that neither single nor double mutations structurally perturbed recombinant proteins consisting of the FH C-terminal modules, 19 and 20 (FH19-20), although all three FH19-20 mutants were poor, compared to wild-type FH19-20, at promoting hemolysis of C3b-coated erythrocytes through competition with full-length FH. Indeed, our new crystal structure of the S1191L mutant of FH19-20 complexed with an activation-specific complement fragment, C3d, was nearly identical to that of the wild-type FH19-20:C3d complex, consistent with mutants binding to C3b with wild-type-like affinity. The S1191L mutation enhanced thermal stability of module 20, whereas the V1197A mutation dramatically decreased it. Thus, although mutant proteins were folded at 37 °C, they differ in conformational rigidity. Neither single substitutions nor double substitutions increased measurably the extent of FH19-20 self-association, nor did these mutations significantly affect the affinity of FH19-20 for three glycosaminoglycans, despite critical roles of module 20 in recognizing polyanionic self-surface markers. Unexpectedly, FH19-20 mutants containing Leu1191 self-associated on a heparin-coated surface to a higher degree than on surfaces coated with dermatan or chondroitin sulfates. Thus, potentially disease-related functional distinctions between mutants, and between FH and FH-related 1, may manifest in the presence of specific glycosaminoglycans.


Subject(s)
Complement Factor H/chemistry , Complement Factor H/genetics , Gene Conversion , Complement C3b/chemistry , Complement C3d/chemistry , Complement Factor H/metabolism , Crystallography, X-Ray , Humans , Mutation , Pichia/genetics , Pichia/metabolism , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Temperature
16.
Mol Immunol ; 48(15-16): 1844-50, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21683447

ABSTRACT

The complement system is a component of innate immunity and is activated by a cascade of protein interactions whose function is vital to our ability to fight infection. When proper regulation fails, the complement system is unable to recognize "self" from "nonself" and, therefore, attacks own tissues leading to autoimmune diseases. The central protein of the complement system is C3, which is the convergence point of three independently activated but communicating pathways. Regulation of C3 occurs through modular proteins which consist of many repeats of complement control protein (CCP) modules. CCP modules have diverse sequences, similar structures, and diverse physicochemical compositions, with excess of charge being a predominant characteristic. The goal of our study is to understand the electrostatic mechanism that underlies the interaction between the C3d domain of C3 and the fourth module of the complement regulator Factor H (FH4). We have performed a computational alanine scan in which we have replaced every ionizable amino acid, one at a time, with an alanine to generate a family of mutants for the C3d-FH4 complex. We have used Poisson-Boltzmann electrostatic calculations in combination with clustering of spatial distributions of electrostatic potentials and free energy calculations to delineate the contribution of each replaced amino acid to the C3d-FH4 interaction. We have analyzed our data in view of a two-step model which separates association into long-range recognition and short-range binding and we have identified key amino acids that contribute to association. We discuss the complex role of C3d in binding FH4 and the bacterial proteins Efb/Ehp from Staphylococcus aureus.


Subject(s)
Complement C3d/chemistry , Complement Factor H/chemistry , Models, Molecular , Alanine/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Complement C3d/metabolism , Complement Factor H/metabolism , Protein Binding , Protein Structure, Tertiary , Static Electricity
17.
J Mol Graph Model ; 29(8): 1047-55, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21605993

ABSTRACT

Factor H (FH) contributes to the regulation of the complement system by binding to polyanionic surfaces and the proteins C3b/C3c/C3d. This implicates charge and electrostatic interactions in recognition and binding of FH. Despite the large amount of experimental and pathology data the exact mechanism at molecular level is not yet known. We have implemented a computational framework for comparative analysis of the charge and electrostatic diversity of FH modules and C3b domains to identify electrostatic hotspots and predict potential binding sites. Our electrostatic potential clustering analysis shows that charge distributions and electrostatic potential distributions are more useful in understanding C3b-FH interactions than net charges alone. We present a model of non-specific electrostatic interactions of FH with polyanion-rich surfaces and specific interactions with C3b, using our computational data and existing experimental data. We discuss the electrostatic contributions to the formation of the C3b-FH complex and the competition between FH and Factor Bb (Bb) for binding to C3b. We also discuss the significance of mutations of charged amino acids in the pathobiology of FH-mediated disease, such as age-related macular degeneration, atypical hemolytic uremic syndrome, and dense deposit disease. Our data can be used to guide future experimental studies.


Subject(s)
Complement Factor H/chemistry , Complement Factor H/metabolism , Amino Acid Substitution , Atypical Hemolytic Uremic Syndrome , Binding Sites , Complement C3b/chemistry , Complement C3b/metabolism , Complement C3c/chemistry , Complement C3c/metabolism , Complement C3d/chemistry , Complement C3d/metabolism , Complement Factor H/genetics , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/metabolism , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/metabolism , Macular Degeneration/genetics , Macular Degeneration/metabolism , Models, Molecular , Polyelectrolytes , Polymers , Protein Binding/physiology , Protein Structure, Tertiary , Static Electricity
18.
Science ; 332(6029): 608-11, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21527715

ABSTRACT

The interaction of complement receptor 2 (CR2)--which is present on B cells and follicular dendritic cells--with its antigen-bound ligand C3d results in an enhanced antibody response, thus providing an important link between the innate and adaptive immune systems. Although a cocrystal structure of a complex between C3d and the ligand-binding domains of CR2 has been published, several aspects of this structure, including the position in C3d of the binding interface, remained controversial because of disagreement with biochemical data. We now report a cocrystal structure of a CR2(SCR1-2):C3d complex at 3.2 angstrom resolution in which the interaction interfaces differ markedly from the previously published structure and are consistent with the biochemical data. It is likely that, in the previous structure, the interaction was influenced by the presence of zinc acetate additive in the crystallization buffer, leading to a nonphysiological complex. Detailed knowledge of the binding interface now at hand gives the potential to exploit the interaction in vaccine design or in therapeutics directed against autoreactive B cells.


Subject(s)
Complement C3d/chemistry , Receptors, Complement 3d/chemistry , Binding Sites , Complement C3d/metabolism , Crystallization , Crystallography, X-Ray , Humans , Hydrogen Bonding , Ligands , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Receptors, Complement 3d/genetics , Receptors, Complement 3d/metabolism , Zinc Acetate
19.
Mol Immunol ; 48(4): 452-62, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21055811

ABSTRACT

The structure of the complement-binding domain of Staphylococcus aureus protein Sbi (Sbi-IV) in complex with ligand C3d is presented. The 1.7Å resolution structure reveals the molecular details of the recognition of thioester-containing fragment C3d of the central complement component C3, involving interactions between residues of Sbi-IV helix α2 and the acidic concave surface of C3d. The complex provides a structural basis for the binding preference of Sbi for native C3 over C3b and explains how Sbi-IV inhibits the interaction between C3d and complement receptor 2. A second C3d binding site on Sbi-IV is identified in the crystal structure that is not observed in related S. aureus C3 inhibitors Efb-C and Ehp. This binding mode perhaps hints as to how Sbi-IV, as part of Sbi, forms a C3b-Sbi adduct and causes futile consumption of C3, an extraordinary aspect of Sbi function that is not shared by any other known Staphylococcal complement inhibitor.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Carrier Proteins/chemistry , Carrier Proteins/immunology , Complement C3d/chemistry , Complement C3d/immunology , Staphylococcus aureus/immunology , Amino Acid Sequence , Amino Acids/immunology , Crystallography, X-Ray , Humans , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Scattering, Small Angle , Structure-Activity Relationship , Surface Properties , Titrimetry
20.
J Mol Biol ; 404(4): 697-710, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20951140

ABSTRACT

The interactions between the complement receptor type 2 (CR2) and the C3 complement fragments C3d, C3dg, and iC3b are essential for the initiation of a normal immune response. A crystal-derived structure of the two N-terminal short consensus repeat (SCR1-2) domains of CR2 in complex with C3d has previously been elucidated. However, a number of biochemical and biophysical studies targeting both CR2 and C3d appear to be in conflict with these structural data. Previous mutagenesis and heteronuclear NMR spectroscopy studies directed toward the C3d-binding site on CR2 have indicated that the CR2-C3d cocrystal structure may represent an encounter/intermediate or nonphysiological complex. With regard to the CR2-binding site on C3d, mutagenesis studies by Isenman and coworkers [Isenman, D. E., Leung, E., Mackay, J. D., Bagby, S. & van den Elsen, J. M. H. (2010). Mutational analyses reveal that the staphylococcal immune evasion molecule Sbi and complement receptor 2 (CR2) share overlapping contact residues on C3d: Implications for the controversy regarding the CR2/C3d cocrystal structure. J. Immunol. 184, 1946-1955] have implicated an electronegative "concave" surface on C3d in the binding process. This surface is discrete from the CR2-C3d interface identified in the crystal structure. We generated a total of 18 mutations targeting the two (X-ray crystallographic- and mutagenesis-based) proposed CR2 SCR1-2 binding sites on C3d. Using ELISA analyses, we were able to assess binding of mutant forms of C3d to CR2. Mutations directed toward the concave surface of C3d result in substantially compromised CR2 binding. By contrast, targeting the CR2-C3d interface identified in the cocrystal structure and the surrounding area results in significantly lower levels of disruption in binding. Molecular modeling approaches used to investigate disparities between the biochemical data and the X-ray structure of the CR2-C3d cocrystal result in highest-scoring solutions in which CR2 SCR1-2 is docked within the concave surface of C3d.


Subject(s)
Complement C3d/chemistry , Complement C3d/genetics , Receptors, Complement 3d/chemistry , Receptors, Complement 3d/metabolism , Amino Acid Substitution , Animals , Binding Sites , Complement C3d/metabolism , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay/methods , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...