Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
J Biol Chem ; 300(7): 107451, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38844131

ABSTRACT

Complement receptor 1 (CR1) is a membrane glycoprotein with a highly duplicated domain structure able to bind multiple ligands such as C3b and C4b, the activated fragments of complement components C3 and C4, respectively. We have previously used our knowledge of this domain structure to identify CSL040, a soluble extracellular fragment of CR1 containing the long homologous repeat (LHR) domains A, B, and C. CSL040 retains the ability to bind both C3b and C4b but is also a more potent complement inhibitor than other recombinant CR1-based therapeutics. To generate soluble CR1 variants with increased inhibitory potential across all three complement pathways, or variants with activity skewed to specific pathways, we exploited the domain structure of CR1 further by generating LHR domain duplications. We identified LHR-ABCC, a soluble CR1 variant containing a duplicated C3b-binding C-terminal LHR-C domain that exhibited significantly enhanced alternative pathway inhibitory activity in vitro compared to CSL040. Another variant, LHR-BBCC, containing duplications of both LHR-B and LHR-C with four C3b binding sites, was shown to have reduced classical/lectin pathway inhibitory activity compared to CSL040, but comparable alternative pathway activity. Interestingly, multiplication of the C4b-binding LHR-A domain resulted in only minor increases in classical/lectin pathway inhibitory activity. The CR1 duplication variants characterized in these in vitro potency assays, as well as in affinity in solution C3b and C4b binding assays, not only provides an opportunity to identify new therapeutic molecules but also additional mechanistic insights to the multiple interactions between CR1 and C3b/C4b.


Subject(s)
Complement C3b , Protein Domains , Humans , Complement C3b/metabolism , Complement C3b/chemistry , Complement C3b/genetics , Receptors, Complement 3b/metabolism , Receptors, Complement 3b/genetics , Receptors, Complement 3b/chemistry , Complement C4b/metabolism , Complement C4b/genetics , Complement C4b/chemistry , Protein Binding
2.
J Biol Chem ; 300(7): 107452, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38852887

ABSTRACT

Rare variants (RVs) in the gene encoding the regulatory enzyme complement factor I (CFI; FI) that reduce protein function or levels increase age-related macular degeneration risk. A total of 3357 subjects underwent screening in the SCOPE natural history study for geographic atrophy secondary to age-related macular degeneration, including CFI sequencing and serum FI measurement. Eleven CFI RV genotypes that were challenging to categorize as type I (low serum level) or type II (normal serum level, reduced enzymatic function) were characterized in the context of pure FI protein in C3b and C4b fluid phase cleavage assays and a novel bead-based functional assay (BBFA) of C3b cleavage. Four variants predicted or previously characterized as benign were analyzed by BBFA for comparison. In all, three variants (W51S, C67R, and I370T) resulted in low expression. Furthermore, four variants (P64L, R339Q, G527V, and P528T) were identified as being highly deleterious with IC50s for C3b breakdown >1 log increased versus the WT protein, while two variants (K476E and R474Q) were ∼1 log reduced in function. Meanwhile, six variants (P50A, T203I, K441R, E548Q, P553S, and S570T) had IC50s similar to WT. Odds ratios and BBFA IC50s were positively correlated (r = 0.76, p < 0.01), while odds ratios versus combined annotation dependent depletion (CADD) scores were not (r = 0.43, p = 0.16). Overall, 15 CFI RVs were functionally characterized which may aid future patient stratification for complement-targeted therapies. Pure protein in vitro analysis remains the gold standard for determining the functional consequence of CFI RVs.


Subject(s)
Complement C3b , Complement Factor I , Genotype , Geographic Atrophy , Humans , Complement Factor I/genetics , Complement Factor I/metabolism , Geographic Atrophy/genetics , Geographic Atrophy/blood , Geographic Atrophy/metabolism , Female , Male , Complement C3b/metabolism , Complement C3b/genetics , Aged , Cohort Studies , Macular Degeneration/genetics , Macular Degeneration/metabolism , Middle Aged
3.
Front Immunol ; 13: 1028760, 2022.
Article in English | MEDLINE | ID: mdl-36643920

ABSTRACT

Age-related macular degeneration (AMD) is linked to 2 main disparate genetic pathways: a chromosome 10 risk locus and the alternative pathway (AP) of complement. Rare genetic variants in complement factor H (CFH; FH) and factor I (CFI; FI) are associated with AMD. FH acts as a soluble cofactor to facilitate FI's cleavage and inactivation of the central molecule of the AP, C3b. For personalised treatment, sensitive assays are required to define the functional significance of individual AP genetic variants. Generation of recombinant FI for functional analysis has thus far been constrained by incomplete processing resulting in a preparation of active and inactive protein. Using an internal ribosomal entry site (IRES)-Furin-CFI expression vector, fully processed FI was generated with activity equivalent to serum purified FI. By generating FI with an inactivated serine protease domain (S525A FI), a real-time surface plasmon resonance assay of C3b:FH:FI complex formation for characterising variants in CFH and CFI was developed and correlated well with standard assays. Using these methods, we further demonstrate that patient-associated rare genetic variants lacking enzymatic activity (e.g. CFI I340T) may competitively inhibit the wild-type FI protein. The dominant negative effect identified in inactive factor I variants could impact on the pharmacological replacement of FI currently being investigated for the treatment of dry AMD.


Subject(s)
Complement C3b , Complement Factor H , Complement Factor I , Macular Degeneration , Humans , Complement C3b/genetics , Macular Degeneration/genetics , Complement Factor H/genetics , Complement Factor I/genetics
4.
J Biol Chem ; 295(48): 16342-16358, 2020 11 27.
Article in English | MEDLINE | ID: mdl-32928961

ABSTRACT

The human complement Factor H-related 5 protein (FHR5) antagonizes the main circulating complement regulator Factor H, resulting in the deregulation of complement activation. FHR5 normally contains nine short complement regulator (SCR) domains, but a FHR5 mutant has been identified with a duplicated N-terminal SCR-1/2 domain pair that causes CFHR5 nephropathy. To understand how this duplication causes disease, we characterized the solution structure of native FHR5 by analytical ultracentrifugation and small-angle X-ray scattering. Sedimentation velocity and X-ray scattering indicated that FHR5 was dimeric, with a radius of gyration (Rg ) of 5.5 ± 0.2 nm and a maximum protein length of 20 nm for its 18 domains. This result indicated that FHR5 was even more compact than the main regulator Factor H, which showed an overall length of 26-29 nm for its 20 SCR domains. Atomistic modeling for FHR5 generated a library of 250,000 physically realistic trial arrangements of SCR domains for scattering curve fits. Only compact domain structures in this library fit well to the scattering data, and these structures readily accommodated the extra SCR-1/2 domain pair present in CFHR5 nephropathy. This model indicated that mutant FHR5 can form oligomers that possess additional binding sites for C3b in FHR5. We conclude that the deregulation of complement regulation by the FHR5 mutant can be rationalized by the enhanced binding of FHR5 oligomers to C3b deposited on host cell surfaces. Our FHR5 structures thus explained key features of the mechanism and pathology of CFHR5 nephropathy.


Subject(s)
Complement System Proteins/chemistry , Kidney Diseases , Mutation , Protein Multimerization , Complement C3b/chemistry , Complement C3b/genetics , Complement C3b/metabolism , Complement System Proteins/genetics , Complement System Proteins/metabolism , HEK293 Cells , Humans , Protein Domains
5.
Infect Immun ; 88(8)2020 07 21.
Article in English | MEDLINE | ID: mdl-32513855

ABSTRACT

The serum complement system is a first line of defense against bacterial invaders. Resistance to killing by serum enhances the capacity of Klebsiella pneumoniae to cause infection, but it is an incompletely understood virulence trait. Identifying and characterizing the factors responsible for preventing activation of, and killing by, serum complement could inform new approaches to treatment of K. pneumoniae infections. Here, we used functional genomic profiling to define the genetic basis of complement resistance in four diverse serum-resistant K. pneumoniae strains (NTUH-K2044, B5055, ATCC 43816, and RH201207), and explored their recognition by key complement components. More than 90 genes contributed to resistance in one or more strains, but only three, rfaH, lpp, and arnD, were common to all four strains. Deletion of the antiterminator rfaH, which controls the expression of capsule and O side chains, resulted in dramatic complement resistance reductions in all strains. The murein lipoprotein gene lpp promoted capsule retention through a mechanism dependent on its C-terminal lysine residue; its deletion led to modest reductions in complement resistance. Binding experiments with the complement components C3b and C5b-9 showed that the underlying mechanism of evasion varied in the four strains: B5055 and NTUH-K2044 appeared to bypass recognition by complement entirely, while ATCC 43816 and RH201207 were able to resist killing despite being associated with substantial levels of C5b-9. All rfaH and lpp mutants bound C3b and C5b-9 in large quantities. Our findings show that, even among this small selection of isolates, K. pneumoniae adopts differing mechanisms and utilizes distinct gene sets to avoid complement attack.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Carboxy-Lyases/immunology , Gene Expression Regulation, Bacterial/immunology , Genes, Bacterial , Immune Evasion , Klebsiella pneumoniae/immunology , Peptide Elongation Factors/immunology , Bacterial Outer Membrane Proteins/genetics , Blood Bactericidal Activity/immunology , Carboxy-Lyases/deficiency , Carboxy-Lyases/genetics , Complement C3b/genetics , Complement C3b/immunology , Complement Membrane Attack Complex/genetics , Complement Membrane Attack Complex/immunology , DNA Transposable Elements , Gene Expression Profiling , Gene Library , Humans , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/pathogenicity , Mutation , Peptide Elongation Factors/deficiency , Peptide Elongation Factors/genetics , Sequence Analysis, DNA
6.
Front Immunol ; 11: 774, 2020.
Article in English | MEDLINE | ID: mdl-32431705

ABSTRACT

We aimed at establishing a sensitive and robust assay for estimation of systemic complement activation at complement component C3 level in mouse and human plasma samples. In order to capture the activation products iC3b and C3dg in a specific and physiological relevant manner we utilized a construct consisting of the iC3b/C3dg-binding site of human complement receptor 2 (CR2) attached to an Fc-part of mouse IgG. This construct binds C3dg and iC3b from both mice and humans. We purified the CR2-IgG construct from mouse B myeloma cell line supernatants, J558L-CR2-IgG, by protein G affinity chromatography. The CR2-IgG construct was used for capturing C3 fragments in microtiter wells and an anti-mouse or an anti-human-C3 antibody was used for detection of bound C3 fragments. Initially we tested the specificity of the assays with the use of purified C3 fragments. Further, with the use of the CR2-based assay, we measured an up to three-fold higher signal in activated mouse serum as compared to non-activated mouse serum, whereas activated serum from a C3 knock-out mouse gave no signal. We tested in vivo generated samples from a mouse experiment; complement activation was induced by injecting cobra venom factor or heat aggregated IgG into C57bl6 mice, followed by withdrawal of EDTA blood samples at different time points and measurement of iC3b/C3dg. We observed a clear time-dependent distinction in signals between samples with expected high and low complement activation. Furthermore, with the use of the assay for human C3 fragments, we observed that patients with systemic lupus erythematosus (SLE) (n = 144) had significantly higher iC3b/C3dg levels as compared to healthy individuals (n = 144) (p < 0.0001). We present two functional immunoassays, that are able to measure systemic levels of the C3-activation products iC3b and C3dg in mice and humans. To our knowledge, these are the first assays for complement activation that use a physiological relevant capture construct such as CR2. These assays will be a relevant tool when investigating mouse models and human diseases involving the complement system.


Subject(s)
Complement Activation/drug effects , Complement C3b/immunology , Fluoroimmunoassay/methods , Lupus Erythematosus, Systemic/blood , Peptide Fragments/immunology , Receptors, Complement 3d/immunology , Animals , Binding Sites/immunology , Cell Line, Tumor , Cohort Studies , Complement C3b/genetics , Cross-Sectional Studies , Elapid Venoms/pharmacology , Gene Knockout Techniques , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Lupus Erythematosus, Systemic/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/genetics
7.
Front Immunol ; 10: 2097, 2019.
Article in English | MEDLINE | ID: mdl-31552043

ABSTRACT

Properdin enhances complement-mediated opsonization of targeted cells and particles for immune clearance. Properdin occurs as dimers, trimers and tetramers in human plasma, which recognize C3b-deposited surfaces, promote formation, and prolong the lifetime of C3bBb-enzyme complexes that convert C3 into C3b, thereby enhancing the complement-amplification loop. Here, we report crystal structures of monomerized properdin, which was produced by co-expression of separate N- and C-terminal constructs that yielded monomer-sized properdin complexes that stabilized C3bBb. Consistent with previous low-resolution X-ray and EM data, the crystal structures revealed ring-shaped arrangements that are formed by interactions between thrombospondin type-I repeat (TSR) domains 4 and 6 of one protomer interacting with the N-terminal domain (which adopts a short transforming-growth factor B binding protein-like fold) and domain TSR1 of a second protomer, respectively. Next, a structure of monomerized properdin in complex with the C-terminal domain of C3b showed that properdin-domain TSR5 binds along the C-terminal α-helix of C3b, while two loops, one from domain TSR5 and one from TSR6, extend and fold around the C3b C-terminus like stirrups. This suggests a mechanistic model in which these TSR5 and TSR6 "stirrups" bridge interactions between C3b and factor B or its fragment Bb, and thereby enhance formation of C3bB pro-convertases and stabilize C3bBb convertases. In addition, properdin TSR6 would sterically block binding of the protease factor I to C3b, thus limiting C3b proteolytic degradation. The presence of a valine instead of a third tryptophan in the canonical Trp-ladder of TSR domains in TSR4 allows a remarkable ca. 60°-domain bending motion of TSR4. Together with variable positioning of TSR2 and, putatively, TSR3, this explains the conformational flexibility required for properdin to form dimers, trimers, and tetramers. In conclusion, the results indicate that binding avidity of oligomeric properdin is needed to distinguish surface-deposited C3b molecules from soluble C3b or C3 and suggest that properdin-mediated interactions bridging C3b-B and C3b-Bb enhance affinity, thus promoting convertase formation and stabilization. These mechanisms explain the enhancement of complement-mediated opsonization of targeted cells and particle for immune clearance.


Subject(s)
Complement Activation , Complement C3b/chemistry , Immunologic Factors/chemistry , Properdin/chemistry , Complement C3b/genetics , Complement C3b/immunology , Glycosylation , HEK293 Cells , Humans , Immunologic Factors/immunology , Properdin/genetics , Properdin/immunology , Protein Domains , Recombinant Proteins/chemistry
8.
Int J Biol Macromol ; 133: 391-411, 2019 Jul 15.
Article in English | MEDLINE | ID: mdl-30974145

ABSTRACT

Previously, we have identified the C3dg protein as an important player in the pathogenesis of atopic dermatitis (AD). In this study, we aimed to identify critical factors associated with C3dg in human keratinocytes based on high-throughput screening (HTS) approaches. We overexpressed C3dg in HaCaT human keratinocytes and conducted serial HTS studies, including RNA sequencing analysis integrated with antibody-chip arrays and implementation of bioinformatics algorithms (PPI mappings). Cumulatively, these approaches identified several novel C3dg-associated genes and proteins that are thought to be significantly involved in skin diseases including AD. These novel genes and proteins included LPA, PROZ, BLK, CLDN11, and FGF22, which are believed to play important roles in C3dg-associated skin functions in keratinocytes, as well as genes related to the two important pathways of systemic lupus erythematosus and Staphylococcus aureus infection. In particular, FGF22 is a unique gene that was detected and validated in all methods applied in this study. By integrating the datasets obtained from these HTS studies and utilizing the strengths of each method, we obtained new insights into the functional role of C3dg in keratinocytes. The approach used here contributes to clinical understanding of C3dg-associated applications and may also be applicable to treatment of AD.


Subject(s)
Antibodies/metabolism , Complement C3b/genetics , Complement C3b/metabolism , Computational Biology , Keratinocytes/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Array Analysis , Sequence Analysis, RNA , Algorithms , Hep G2 Cells , Humans
9.
J Biol Chem ; 293(44): 17166-17187, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30217822

ABSTRACT

Factor H (FH) is the major regulator of C3b in the alternative pathway of the complement system in immunity. FH comprises 20 short complement regulator (SCR) domains, including eight glycans, and its Y402H polymorphism predisposes those who carry it to age-related macular degeneration. To better understand FH complement binding and self-association, we have studied the solution structures of both the His-402 and Tyr-402 FH allotypes. Analytical ultracentrifugation revealed that up to 12% of both FH allotypes self-associate, and this was confirmed by small-angle X-ray scattering (SAXS), MS, and surface plasmon resonance analyses. SAXS showed that monomeric FH has a radius of gyration (Rg ) of 7.2-7.8 nm and a length of 25 nm. Starting from known structures for the SCR domains and glycans, the SAXS data were fitted using Monte Carlo methods to determine atomistic structures of monomeric FH. The analysis of 29,715 physically realistic but randomized FH conformations resulted in 100 similar best-fit FH structures for each allotype. Two distinct molecular structures resulted that showed either an extended N-terminal domain arrangement with a folded-back C terminus or an extended C terminus and a folded-back N terminus. These two structures are the most accurate to date for glycosylated full-length FH. To clarify FH functional roles in host protection, crystal structures for the FH complexes with C3b and C3dg revealed that the extended N-terminal conformation accounted for C3b fluid-phase regulation, the extended C-terminal conformation accounted for C3d binding, and both conformations accounted for bivalent FH binding to glycosaminoglycans on the target cell surface.


Subject(s)
Complement C3b , Complement Factor H , Peptide Fragments , Complement C3b/chemistry , Complement C3b/genetics , Complement C3b/metabolism , Complement Factor H/chemistry , Complement Factor H/genetics , Complement Factor H/metabolism , Crystallography, X-Ray , Humans , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Protein Conformation , Protein Domains , Surface Plasmon Resonance , X-Ray Diffraction
10.
Free Radic Biol Med ; 129: 237-246, 2018 12.
Article in English | MEDLINE | ID: mdl-30253188

ABSTRACT

Complement Factor H (CFH) is an important inhibitor of the alternate complement pathway in Bruch's membrane (BM), located between the choriocapillaris and the retinal pigment epithelium. Furthermore dysfunction of its activity as occurs with certain polymorphisms is associated with an increased risk of age related macular degeneration (AMD). The retina is a site of high generation of reactive oxygen species (ROS) and dysfunction of redox homeostasis in this milieu also contributes to AMD pathogenesis. In this study we wanted to explore if CFH exists in distinct redox forms and whether these species have unique protective biological functions. CFH can be reduced by the naturally occurring thioredoxin - 1 in CFH domains 1-4, 17-20. We found a duality of function between the oxidised and reduced forms of CFH. The oxidised form was more efficient in binding to C3b and lipid peroxidation by-products that are known to accumulate in the retinae and activate the alternate complement pathway. Oxidised CFH enhances Factor I mediated cleavage of C3 and C3b whereas the reduced form loses this activity. In the setting of oxidative stress (hydrogen peroxide)-mediated death of human retinal pigment epithelial cells as can occur in AMD, the free thiol form of CFH offers a protective function compared to the oxidised form. We found for the first time using a novel ELISA system we have developed for free thiol CFH, that both redox forms of CFH are found in the human plasma. Furthermore there is a distinct ratio of these redox forms in plasma depending if an individual has early or late AMD, with individuals with early AMD having higher levels of the free thiol form compared to late AMD.


Subject(s)
Complement C3b/metabolism , Complement Factor I/metabolism , Macular Degeneration/genetics , Reactive Oxygen Species/metabolism , Aged , Bruch Membrane/immunology , Bruch Membrane/pathology , Case-Control Studies , Cell Line , Complement Activation/genetics , Complement C3b/genetics , Complement Factor H/genetics , Complement Factor H/metabolism , Complement Factor I/genetics , Complement Pathway, Alternative/genetics , Epithelial Cells/cytology , Epithelial Cells/immunology , Female , Gene Expression , Humans , Lipid Peroxidation , Macular Degeneration/immunology , Macular Degeneration/pathology , Male , Oxidation-Reduction , Protein Binding , Proteolysis , Reactive Oxygen Species/immunology , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/pathology , Time Factors
11.
Hum Genet ; 137(4): 305-314, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29675612

ABSTRACT

Single nucleotide variants (SNVs) within and surrounding the complement receptor 1 (CR1) gene show some of the strongest genome-wide association signals with late-onset Alzheimer's disease. Some studies have suggested that this association signal is due to a duplication allele (CR1-B) of a low copy repeat (LCR) within the CR1 gene, which increases the number of complement C3b/C4b-binding sites in the mature receptor. In this study, we develop a triplex paralogue ratio test assay for CR1 LCR copy number allowing large numbers of samples to be typed with a limited amount of DNA. We also develop a CR1-B allele-specific PCR based on the junction generated by an historical non-allelic homologous recombination event between CR1 LCRs. We use these methods to genotype CR1 and measure CR1-B allele frequency in both late-onset and early-onset cases and unaffected controls from the United Kingdom. Our data support an association of late-onset Alzheimer's disease with the CR1-B allele, and confirm that this allele occurs most frequently on the risk haplotype defined by SNV alleles. Furthermore, regression models incorporating CR1-B genotype provide a better fit to our data compared to incorporating the SNV-defined risk haplotype, supporting the CR1-B allele as the variant underlying the increased risk of late-onset Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , DNA Copy Number Variations/genetics , Genetic Predisposition to Disease , Receptors, Complement 3b/genetics , Adult , Aged , Aged, 80 and over , Alleles , Alzheimer Disease/physiopathology , Binding Sites , Complement C3b/genetics , Complement C4b-Binding Protein/genetics , Female , Gene Duplication/genetics , Genome-Wide Association Study , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Risk Factors , United Kingdom
12.
Dev Comp Immunol ; 84: 250-263, 2018 07.
Article in English | MEDLINE | ID: mdl-29501422

ABSTRACT

The evolution of complement system in invertebrates is poorly investigated. While the repertoire of complement genes in several Ecdysozoa lineages is found substantially different from that of Deuterostomia, the composition and function of the complement in the second protostome lineage, Lophotrochozoa, remains unclear. Here we report the general description of new transcriptomic data on the common periwinkle, Littorina littorea, and trace the evolutionary trajectories of the ancestral proto-complement repertoire. The repertoire is defined as immune cascade providing the minimum set of C3-associated molecules required for C3b amplification, opsonization of the targets and their phagocytosis: thioester protein (TEP) C3, serine protease C2/factor B (Bf) and complement receptors (CR). The reference transcriptome of L. littorea was built from the dual-species RNA-seq experiment with the periwinkle and its tissue digenean parasite Himasthla elongata. Five TEPs, including the ortholog of the C3, are found expressed in the in the mollusk's inflamed tissues. The homolog of the complement receptors CR1/CR2 is also expressed, however the ortholog of Bf is not. The extensive phylogenetic analysis showed that the C3 ortholog and the complement receptors are retained in all key lophotrochozoan taxa: Mollusca, Annelida and Brachiopoda. However, the Bf ortholog was lost at least three times independently in different lineages: i) Cephalopoda, ii) a common ancestor of all Gastropoda and iii) one of the Annelida lineage, Clitellata. Both C3 and Bf molecules were retained in bivalve species, brachiopods and annelid worms from the Polychaeta lineage. Hypothetically, the function of the lost Bf in these animals can be compensated by Factor L (Lf) - the serine protease first found in L. littorea and homologous to both, the Bf and the arthropod factor C (Cf). The contrast differences in proto-complement repertoire between the sister mollusk' taxa, Bivalvia and Gastropoda (the conserved and modified sets, respectively), can underlie differences in their susceptibility to digenean infection.


Subject(s)
Complement C3b/genetics , Gastropoda/physiology , Transcriptome/immunology , Trematoda/physiology , Trematode Infections/genetics , Animals , Annelida , Biological Evolution , Cephalopoda , Complement Activation/genetics , Complement Factor B/genetics , Gene Expression Profiling , Host-Parasite Interactions , Humans , Immunity, Innate , Inflammation/genetics , Invertebrates , Phagocytosis/genetics , Phylogeny , Receptors, Complement/genetics
13.
Mol Immunol ; 85: 137-147, 2017 05.
Article in English | MEDLINE | ID: mdl-28254726

ABSTRACT

C3b, the central component of the alternative pathway (AP) of the complement system, coexists as a mixture of conformations in solution. These conformational changes can affect interactions with other proteins and complement regulators. Here we combine a computational model for electrostatic interactions within C3b with molecular imaging to study the conformation of C3b. The computational analysis shows that the TED domain in C3b is tethered ionically to the macroglobulin (MG) ring. Monovalent counterion concentration affects the magnitude of electrostatic forces anchoring the TED domain to the rest of the C3b molecule in a thermodynamic model. This is confirmed by observing NaCl concentration dependent conformational changes using single molecule electron microscopy (EM). We show that the displacement of the TED domain is compatible with C3b binding to Factor B (FB), suggesting that the regulation of the C3bBb convertase could be affected by conditions that promote movement in the TED domain. Our molecular model also predicts mutations that could alter the positioning of the TED domain, including the common R102G polymorphism, a risk variant for developing age-related macular degeneration. The common C3b isoform, C3bS, and the risk isoform, C3bF, show distinct energetic barriers to displacement in the TED that are related to a network of electrostatic interactions at the interface of the TED and MG-ring domains of C3b. These computational predictions agree with experimental evidence that shows differences in conformation observed in C3b isoforms purified from homozygous donors. Altogether, we reveal an ionic, reversible attachment of the TED domain to the MG ring that may influence complement regulation in some mutations and polymorphisms of C3b.


Subject(s)
Complement C3b/chemistry , Complement C3b/metabolism , Macular Degeneration/genetics , Models, Molecular , Animals , Complement C3b/genetics , Genetic Predisposition to Disease , Humans , Microscopy, Electron , Polymorphism, Single Nucleotide , Protein Conformation , Protein Domains/physiology , Protein Stability , Thermodynamics
14.
Proc Natl Acad Sci U S A ; 114(13): 3403-3408, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28292891

ABSTRACT

Recognition by the leukocyte integrins αXß2 and αMß2 of complement iC3b-opsonized targets is essential for effector functions including phagocytosis. The integrin-binding sites on iC3b remain incompletely characterized. Here, we describe negative-stain electron microscopy and biochemical studies of αXß2 and αMß2 in complex with iC3b. Despite high homology, the two integrins bind iC3b at multiple distinct sites. αXß2 uses the αX αI domain to bind iC3b on its C3c moiety at one of two sites: a major site at the interface between macroglobulin (MG) 3 and MG4 domains, and a less frequently used site near the C345C domain. In contrast, αMß2 uses its αI domain to bind iC3b at the thioester domain and simultaneously interacts through a region near the αM ß-propeller and ß2 ßI domain with a region of the C3c moiety near the C345C domain. Remarkably, there is no overlap between the primary binding site of αXß2 and the binding site of αMß2 on iC3b. Distinctive binding sites on iC3b by integrins αXß2 and αMß2 may be biologically beneficial for leukocytes to more efficiently capture opsonized pathogens and to avoid subversion by pathogen factors.


Subject(s)
Complement C3b/metabolism , Integrin alphaXbeta2/metabolism , Macrophage-1 Antigen/metabolism , Binding Sites , Complement C3b/chemistry , Complement C3b/genetics , Humans , Integrin alphaXbeta2/chemistry , Integrin alphaXbeta2/genetics , Leukocytes/chemistry , Leukocytes/metabolism , Macrophage-1 Antigen/chemistry , Macrophage-1 Antigen/genetics , Protein Binding , Protein Domains , Protein Structure, Tertiary
15.
JAMA Ophthalmol ; 135(1): 39-46, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27918759

ABSTRACT

IMPORTANCE: In age-related macular degeneration (AMD), rare variants in the complement system have been described, but their functional consequences remain largely unexplored. OBJECTIVES: To identify new rare variants in complement genes and determine the functional effect of identified variants on complement levels and complement regulation in serum samples from carriers and noncarriers. DESIGN, SETTING, AND PARTICIPANTS: This study evaluated affected (n = 114) and unaffected (n = 60) members of 22 families with AMD and a case-control cohort consisting of 1831 unrelated patients with AMD and 1367 control individuals from the European Genetic Database from March 29, 2006, to April 26, 2013, in Nijmegen, the Netherlands, and Cologne, Germany. Exome sequencing data of families were filtered for rare variants in the complement factor H (CFH), complement factor I (CFI), complement C9 (C9), and complement C3 (C3) genes. The case-control cohort was genotyped with allele-specific assays. Serum samples were obtained from carriers of identified variants (n = 177) and age-matched noncarriers (n = 157). Serum concentrations of factor H (FH), factor I (FI), C9, and C3 were measured, and C3b degradation ability was determined. MAIN OUTCOMES AND MEASURES: Association of rare variants in the CFH, CFI, C9, and C3 genes with AMD, serum levels of corresponding proteins, and C3b degradation ability of CFH and CFI variant carriers. RESULTS: The 1831 unrelated patients with AMD had a mean (SD) age of 75.0 (9.4) years, and 60.5% were female. The 1367 unrelated control participants had a mean (SD) age of 70.4 (7.0), and 58.7% were female. All individuals were of European descent. Rare variants in CFH, CFI, C9, and C3 contributed to an increased risk of developing AMD (odds ratio, 2.04; 95% CI, 1.47-2.82; P < .001). CFI carriers had decreased median FI serum levels (18.2 µg/mL in Gly119Arg carriers and 16.2 µg/mL in Leu131Arg carriers vs 27.2 and 30.4 µg/mL in noncarrier cases and controls, respectively; both P < .001). Elevated C9 levels were observed in Pro167Ser carriers (10.7 µg/mL vs 6.6 and 6.1 µg/mL in noncarrier cases and controls, respectively; P < .001). The median FH serum levels were 299.4 µg/mL for CFH Arg175Gln and 266.3 µg/mL for CFH Ser193Leu carriers vs 302.4 and 283.0 µg/mL for noncarrier cases and controls, respectively. The median C3 serum levels were 943.2 µg/mL for C3 Arg161Trp and 946.7 µg/mL for C3 Lys155Gln carriers vs 874.0 and 946.7 µg/mL for noncarrier cases and controls, respectively. The FH and FI levels correlated with C3b degradation in noncarriers (R2 = 0.35 and R2 = 0.31, respectively; both P < .001). CONCLUSIONS AND RELEVANCE: Reduced serum levels were associated with C3b degradation in carriers of CFI but not CFH variants, suggesting that CFH variants affect functional activity of FH rather than serum levels. Carriers of CFH (Arg175Gln and Ser193Leu) and CFI (Gly119Arg and Leu131Arg) variants have an impaired ability to regulate complement activation and may benefit more from complement-inhibiting therapy than patients with AMD in general.


Subject(s)
Complement C3b/genetics , DNA/genetics , Macular Degeneration/genetics , Aged , Aged, 80 and over , Alleles , Complement C3b/metabolism , Exome , Female , Genotype , Germany/epidemiology , Humans , Macular Degeneration/epidemiology , Macular Degeneration/metabolism , Male , Middle Aged , Netherlands/epidemiology , Polymorphism, Single Nucleotide , Prevalence , Retrospective Studies
16.
Matrix Biol ; 57-58: 299-310, 2017 01.
Article in English | MEDLINE | ID: mdl-27609404

ABSTRACT

The glomerular basement membrane (GBM) is an essential component of the glomerular filtration barrier. Heparan sulfate proteoglycans such as agrin are major components of the GBM, along with α345(IV) collagen, laminin-521 and nidogen. A loss of GBM heparan sulfate chains is associated with proteinuria in several glomerular diseases and may contribute to the underlying pathology. As the major determinants of the anionic charge of the GBM, heparan sulfate chains have been thought to impart charge selectivity to the glomerular filtration, a view challenged by the negligible albuminuria in mice that lack heparan sulfate in the GBM. Recent studies provide increasing evidence that heparan sulfate chains modulate local complement activation by recruiting complement regulatory protein factor H, the major inhibitor of the alternative pathway in plasma. Factor H selectively inactivates C3b bound to surfaces bearing host-specific polyanions such as heparan sulfate, thus limiting complement activation on self surfaces such as the GBM, which are not protected by cell-bound complement regulators. We discuss mechanisms whereby the acquired loss of GBM heparan sulfate can impair the local regulation of the alternative pathway, exacerbating complement activation and glomerular injury in immune-mediated kidney diseases such as membranous nephropathy and lupus nephritis.


Subject(s)
Complement Activation , Gene Expression Regulation/immunology , Glomerular Basement Membrane/immunology , Glomerulonephritis, Membranous/immunology , Heparitin Sulfate/immunology , Lupus Nephritis/immunology , Agrin/genetics , Agrin/immunology , Animals , Collagen Type IV/genetics , Collagen Type IV/immunology , Complement C3b/genetics , Complement C3b/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Glomerular Basement Membrane/metabolism , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/pathology , Heparitin Sulfate/metabolism , Humans , Laminin/genetics , Laminin/immunology , Lupus Nephritis/genetics , Lupus Nephritis/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Signal Transduction , Static Electricity
17.
Clin Transplant ; 31(1)2017 01.
Article in English | MEDLINE | ID: mdl-27801525

ABSTRACT

Complement component 3 (C3) presents both slow (C3S) and fast (C3F) variants, which can be locally produced and activated by immune system cells. We studied C3 recipient variants in 483 liver transplant patients by RT-PCR-HRM to determine their effect on graft outcome during the first year post-transplantation. Allograft survival was significantly decreased in C3FF recipients (C3SS 95% vs C3FS 91% vs C3FF 83%; P=.01) or C3F allele carriers (C3F absence 95% vs C3F presence 90%, P=.02). C3FF genotype or presence of C3F allele independently increased risk for allograft loss (OR: 2.38, P=.005 and OR: 2.66, P=.02, respectively). C3FF genotype was more frequent among patients whose first infection was of viral etiology (C3SS 13% vs C3FS 18% vs C3FF 32%; P=.04) and independently increased risk for post-transplant viral infections (OR: 3.60, P=.008). On the other hand, C3FF and C3F protected from rejection events (OR: 0.54, P=.03 and OR: 0.63, P=.047, respectively). Differences were not observed in hepatitis C virus recurrence or patient survival. In conclusion, we show that, independently from C3 variants produced by donor liver, C3F variant from recipient diminishes allograft survival, increases susceptibility to viral infections, and protects from rejection after transplantation. C3 genotyping of liver recipients may be useful to stratify risk.


Subject(s)
Complement C3b/genetics , Graft Rejection/prevention & control , Liver Transplantation/adverse effects , Polymorphism, Genetic , Tissue Donors , Transplant Recipients , Virus Diseases/etiology , Adolescent , Adult , Aged , Biomarkers/metabolism , Child , Child, Preschool , Female , Follow-Up Studies , Graft Rejection/etiology , Graft Survival , Humans , Infant , Infant, Newborn , Male , Middle Aged , Postoperative Complications , Prognosis , Protein Isoforms , Risk Factors , Transplantation, Homologous , Virus Diseases/pathology , Young Adult
18.
FEBS Lett ; 590(15): 2418-34, 2016 08.
Article in English | MEDLINE | ID: mdl-27393384

ABSTRACT

The alternative pathway (AP) of complement can recognize nonself structures by only two molecules, C3b and factor H. The AP deposits C3b covalently on nonself structures via an amplification system. The actual discrimination is performed by factor H, which has binding sites for polyanions (sialic acids, glycosaminoglycans, phospholipids). This robust recognition of 'self' protects our own intact viable cells and tissues, while activating structures are recognized by default. Foreign targets are opsonized for phagocytosis or killed. Mutations in factor H predispose to severe diseases. In hemolytic uremic syndrome, they promote complement attack against blood cells and vascular endothelial cells and lead, for example, to kidney and brain damage. Even pathogens can exploit factor H. In fact, the ability to bind factor H discriminates most pathogenic microbes from nonpathogenic ones.


Subject(s)
Complement Pathway, Alternative/genetics , Hemolytic-Uremic Syndrome/genetics , Phagocytosis/genetics , Bacteria/genetics , Bacteria/pathogenicity , Binding Sites , Brain Injuries/genetics , Brain Injuries/metabolism , Brain Injuries/pathology , Complement C3b/genetics , Complement C3b/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Glycosaminoglycans/metabolism , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/microbiology , Humans , Mutation , Phospholipids/metabolism , Sialic Acids/metabolism
19.
J Biol Chem ; 291(17): 9181-9, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26945067

ABSTRACT

Evasion of complement-mediated opsonophagocytosis enables group A Streptococcus pyogenes (GAS) to establish infection. Different strain-dependent mechanisms are employed by the host to accomplish this goal. In general, GAS inhibits the amplification of the complement cascade on its cell surface by facilitating the degradation of C3b, an opsonin, to an inactive product, inactivated C3b (iC3b), in a step catalyzed by factor I (FI) and its cofactor, factor H (FH), with or without the participation of human host plasmin (hPm). GAS recruits FH to its cell surface via FH receptors, which are transcriptionally controlled by the two-component cluster of virulence responder-sensor system. The manner in which FI-FH and hPm function together on GAS cells is unknown. Using GAS strain AP53, which strongly binds host human plasminogen/plasmin (hPg/hPm) directly via an hPg/hPm surface receptor (PAM), we show that both FI-FH and hPm sequentially cleave C3b. Whereas FI-FH proteolytically cleaves C3b into iC3b, PAM-bound hPm catalyzes cleavage of iC3b into multiple smaller peptides. Unlike AP53, GAS strain M23ND weakly binds FH and recruits hPg/hPm to its cell surface indirectly via fibrinogen bound to M-protein, M23. In this case, FH-FI cleaves C3b into iC3b, with negligible degradation of iC3b by hPm that is bound to fibrinogen on the cells. AP53 and M23ND display similar resistance to human neutrophil-mediated phagocytosis, which results in a corresponding high lethality in mice after injection of these cells. These results suggest that GAS utilizes diverse mechanisms to degrade C3b and thus to protect bacterial cells from the complement response of the host.


Subject(s)
Complement C3b/immunology , Neutrophils/immunology , Phagocytosis , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Streptococcus pyogenes/pathogenicity , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Carrier Proteins/genetics , Carrier Proteins/immunology , Complement C3b/genetics , Humans , Mice , Mice, Transgenic , Neutrophils/pathology , Species Specificity , Streptococcal Infections/genetics , Streptococcal Infections/pathology , Streptococcus pyogenes/genetics
20.
Clin Exp Immunol ; 183(1): 150-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26415566

ABSTRACT

In this paper we have extended our earlier studies of the action of increasing Factor I concentration on complement activation by using a soluble activator, lipopolysaccharide (LPS) endotoxin, and using human erythrocytes as a source of CR1 - the co-factor needed for the final clip of iC3b to C3dg by Factor I. Using this more physiological system, the results show that we can predict that a quite modest increase in Factor I concentration - 22 µg/ml of extra Factor I - will convert the activity of the highest risk sera to those of the lowest risk. Preliminary experiments have been performed with erythrocytes allotyped for CR1 number. While we have not been able to perform an adequate study of their co-factor activities in our assays, preliminary experiments suggest that when Factor I levels are increased the difference produced by different allotypes of red cells is largely overcome. This suggests that in patients with paroxysmal nocturnal haemoglobinuria (PNH) treated with eculizumab, additional treatment with Factor I may be very useful in reducing the need for blood transfusion. We have also explored the age-related allele frequency for the two polymorphisms of Factor H and the polymorphism of C3. In our population, unlike the 1975 study, we found no age variation in the allele frequency in these polymorphisms. This may, however, reflect that the Cambridge BioResource volunteers do not include many very young or very elderly patients, and in general comprise a population not greatly at risk of death from infectious disease.


Subject(s)
Complement C3b/metabolism , Complement Factor H/genetics , Complement Factor I/immunology , Erythrocytes/immunology , Hemoglobinuria, Paroxysmal/blood , Receptors, Complement 3b/immunology , Adolescent , Adult , Age Factors , Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Complement C3b/genetics , Complement Factor I/analysis , Down-Regulation , Gene Frequency , Hemoglobinuria, Paroxysmal/therapy , Humans , Immune Sera/metabolism , Lipopolysaccharides/immunology , Middle Aged , Polymorphism, Genetic , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL